ArticlePDF Available

Cryopreserved Versus Fresh Peripheral Blood Allogeneic Stem Cell Transplantation Outcomes in Patients Receiving Post-transplant Cyclophosphamide for Graft-versus-host Prophylaxis during the COVID-19 Pandemic: A Single Center Experience

Authors:
  • AdventHealth (formerly Florida Hospital) Cancer Institute

Abstract

Background/objectiveCryopreservation of grafts is not common practice in allogeneic hematopoietic stem cell transplant (HSCT) recipients. However, our center had to use cryopreserved cells for allogeneic HSCT during the COVID-19 pandemic to avoid delays in transplantation due to uncertainty regarding patient and donor exposures.Study designWe retrospectively evaluated post-transplant engraftment and survival outcomes of adult patients who received cryopreserved versus fresh allografts during the COVID-19 pandemic.ResultsFifty-five patients with hematologic malignancies received either cryopreserved (n = 34) or fresh (n = 21) allogeneic HSCT using peripheral blood stem cells between January 2020 and December 2020. At a median follow-up time of 15 months, cryopreserved allograft recipients had significantly lower overall survival (OS) (p = 0.02). They also experienced significantly delayed neutrophil (p = 0.01) and platelet engraftments (p < 0.0001), as well as higher red blood cell transfusion-dependence after day + 60 (67.6% vs. 28.6%; p = 0.01). Significantly more cryopreserved allograft recipients received donor lymphocyte infusion than fresh allograft recipients (35.3% vs. 4.8%, p = 0.01). Neither relapse-free survival nor non-relapse mortality differed significantly between the two groups.Conclusion Cryopreservation of allografts in combination with post-transplant cyclophosphamide may negatively affect engraftment and OS outcomes in HSCT recipients.
Vol.:(0123456789)
1 3
International Journal of Hematology
https://doi.org/10.1007/s12185-022-03493-8
ORIGINAL ARTICLE
Cryopreserved versusfresh peripheral blood allogeneic stem cell
transplantation outcomes inpatients receiving post‑transplant
cyclophosphamide forgraft‑versus‑host prophylaxis
duringtheCOVID‑19 pandemic: asingle center experience
MengniGuo1· JieyingLiu1· PamelaClark2· SarfrazAhmad3· RushangPatel2· JuanCarlosVarela2· ShahramMori2
Received: 21 April 2022 / Revised: 7 November 2022 / Accepted: 7 November 2022
© Japanese Society of Hematology 2022
Abstract
Background/objective Cryopreservation of grafts is not common practice in allogeneic hematopoietic stem cell transplant
(HSCT) recipients. However, our center had to use cryopreserved cells for allogeneic HSCT during the COVID-19 pandemic
to avoid delays in transplantation due to uncertainty regarding patient and donor exposures.
Study design We retrospectively evaluated post-transplant engraftment and survival outcomes of adult patients who received
cryopreserved versus fresh allografts during the COVID-19 pandemic.
Results Fifty-five patients with hematologic malignancies received either cryopreserved (n = 34) or fresh (n = 21) alloge-
neic HSCT using peripheral blood stem cells between January 2020 and December 2020. At a median follow-up time of
15months, cryopreserved allograft recipients had significantly lower overall survival (OS) (p = 0.02). They also experienced
significantly delayed neutrophil (p = 0.01) and platelet engraftments (p < 0.0001), as well as higher red blood cell transfusion-
dependence after day + 60 (67.6% vs. 28.6%; p = 0.01). Significantly more cryopreserved allograft recipients received donor
lymphocyte infusion than fresh allograft recipients (35.3% vs. 4.8%, p = 0.01). Neither relapse-free survival nor non-relapse
mortality differed significantly between the two groups.
Conclusion Cryopreservation of allografts in combination with post-transplant cyclophosphamide may negatively affect
engraftment and OS outcomes in HSCT recipients.
Keywords Cryopreservation· Engraftment· Allogeneic· Transplantation· COVID-19
Introduction
Cryopreservation of hematopoietic progenitor cells (HPCs)
in autologous and cord blood transplantation is a common
practice. On the other hand, fresh allograft infusion is stand-
ard in allogeneic hematopoietic stem cell transplantation
(allo-HSCT) [1].
In the pre-COVID-19 era, cryopreserved allo-HSCT was
typically reserved for donor accommodation, and unantici-
pated changes in patient status. Transplanting cryopreserved
HPCs provides several advantages. It offers the ability to
evaluate HPC quality while ensuring an adequate amount is
collected prior to the initiation of conditioning regimen, and
it provides a solution to logistical challenges such as flex-
ibility of international transportation and other unforeseen
obstacles [2]. These benefits are offset by decreased viability
of cryopreserved cells, the toxic effect of longer term cell
exposure to dimethyl sulfoxide (DMSO), added cost, and
more graft manipulation [3, 4].
Although there is plenty of evidence on the safety and
efficacy of cryopreserved autograft [5], data on the outcomes
of cryopreserved allo-HSCT has been mixed and limited
[214]. Some studies showed increased risk of graft failure
* Shahram Mori
shahram.mori@HCAhealthcare.com
1 Department ofInternal Medicine AdventHealth, Orlando,
FL32804, USA
2 Blood andMarrow Transplant Center, AdventHealth Cancer
Institute, AHMG2415 N. Orange Ave., Suite 601, Orlando,
FL32804, USA
3 Gynecologic Oncology Program, AdventHealth Cancer
Institute, Orlando, FL32804, USA
M.Guo et al.
1 3
and higher mortality rate, while others showed no impact on
survival or engraftment.
Due to travel restrictions imposed by the COVID-19 pan-
demic, transplant centers were faced with the uncertainty
of graft and/or donor transport. To ensure allo-HSCT graft
availability at the scheduled infusion time, the National
Marrow Donor Program® (NMDP) required all unrelated
donor (URD) products to be cryopreserved prior to initia-
tion of conditioning regimen starting on March 30, 2020.
This requirement was relaxed to allow fresh allografts to
be infused for collections scheduled on or after August 10,
2020 [15]. Although cryopreservation was not required for
related donor products, concern over COVID-19 infections
forced many transplant centers to cryopreserve these prod-
ucts as well.
We retrospectively examined the effect of cryopreserva-
tion on the outcomes of allo-HSCT in 2020 at our institu-
tion. The outcomes in 55 adult patients with hematologic
malignancies who underwent either cryopreserved or fresh
allo-HSCT using peripheral blood stem cells (PBSC) as the
graft source between January 2020 and December 2020 were
analyzed.
Materials andmethods
Study population
A single center, retrospective cohort study was conducted.
All adult patients undergoing allo-HSCT for hematologic
malignancies between January 1, 2020 and December 31,
2020 at our transplant center were included in this analy-
sis. All patients received PBSC grafts and were censored at
last documented follow-up visit, with a minimum follow-
up period of 12months in surviving patients. Donors were
tested for SARS-CoV-2 by RT-PCR prior to graft collec-
tion, and recipients were tested for SARS-CoV-2 by RT-PCR
before starting the conditioning regimen. This study was
approved by the AdventHealth Institutional Review Board.
All patients provided written informed consent.
HPC product collection, cryopreservation
procedure, analysis, andinfusion
All allogeneic HPC products were collected via peripheral
blood apheresis from granulocyte-colony stimulating factor
(G-CSF) mobilized donors. Collections took place at our
institution or at other NMDP centers. At our institution,
all collections were conducted on Spectra Optia apheresis
instruments (Terumo BCT, Lakewood CO).The HPC prod-
ucts were then processed (plasma depleted, if required) and
either infused fresh or cryopreserved and thawed prior to
infusion in a 37°C water bath using gentle agitation to a
slushy consistency. All cryopreserved products were mixed
with a freezing media containing Plasmalyte, 5% human
serum albumin (HSA) (Baxter, Deerfield, IL) or concur-
rent plasma and DMSO to achieve a final concentration of
10% DMSO and frozen in a controlled rate freezer (Pla-
nar, Kryo 560, Middlesex, UK) to approximately -90°C,
then transferred to a liquid nitrogen storage freezer (less
than -165°C).All HPC products were frozen within 24h
of collection if collected on-site, within 48h if collected
domestically, and within 72h if collected internationally.
All cryopreserved products were thawed either in the cell
therapy facility or at bedside and were infused within 60min
of thawing. All HPC products were tested for CD34+ and
CD3+, then were calculated and reported as cells × 106/L and
cells × 106/kg of recipient body weight. CD34+concentration
(per µL), frequency (among CD45+events) and viability [fre-
quency of 7-amino-actinomycin D (7-AAD)] were assessed
using the Stem Cell Enumeration (SCE) kit (Becton–Dick-
inson, Heidelberg, Germany) as a single-platform.
Endpoints Definition
Engraftment kinetics, overall survival (OS), and relapse-free
survival (RFS) were evaluated. OS was defined as the time
from transplantation until death. RFS was defined as the time
from transplantation until disease relapse. Other specific sec-
ondary outcomes included neutrophil engraftment, platelet
engraftment, graft failure, poor graft function, cumulative
incidence of acute GVHD (aGVHD) and chronic GVHD
(cGVHD), and non-relapse mortality (NRM). Neutrophil
engraftment was defined as the first of three consecutive
days with an absolute neutrophil count (ANC) > 0.5 × 109/L,
and platelet engraftment was defined as the first of three
consecutive days with platelet count > 20 × 109/L without
transfusions in the prior 7days. Primary graft failure was
defined as never having achieved an ANC 0.5 × 109/L
in the absence of disease relapse. Secondary graft failure
was defined as loss of a previously functioning graft asso-
ciated with loss of full donor chimerism. Poor graft func-
tion was defined as two or three cytopenias for > 2weeks,
after day + 28 in the presence of donor chimerism > 5%
[16]. Acute and chronic GVHD were diagnosed and graded
according to standard criteria [17, 18]. Complete remission
(CR) was defined by standard morphological criteria. Liquid
transit time was defined as the time from completion of stem
cell collection to start of stem cell cryopreservation. Time
to infusion was defined as the time from completion of stem
cell collection to completion of stem cell infusion.
Statistical analysis
We used the Kaplan–Meier analysis to compare the OS, RFS,
and NRM of the patients transplanted with cryopreserved or
Cryopreserved versusfresh peripheral blood allogeneic stem cell transplantation outcomes…
1 3
fresh stem cells. We plotted accumulated incidence for the
time to neutrophil engraftment and platelet engraftment.
Univariate linear regression was used to evaluate the corre-
lation between liquid transit time and engraftment. We used
log-rank test for OS, RFS, NRM, and time to engraftment.
Censor date was set at March 20, 2022. The Cox propor-
tional hazards model was used for the multivariant analysis
for the OS, RFS, and NRM. The multivariant analysis of
OS, RFS, and NRM included type of transplantation (cryo-
preserved vs. fresh), age, disease status at the time of trans-
plant (CR vs. non-CR), moderate to severe cGVHD, and
CMV reactivation by day 100. We used the Mann–Whitney
U test to compare the continuous variables. Chi square test
and Fisher’s exact test were used to compare the categorical
variables between patients who had cryopreserved or fresh
transplantation. All analyses were done in R version 4.0.
Results
Patient andgraft baseline characteristics
A total of 55 patients who underwent allo-HSCT in the year
of 2020 were analyzed in the study. Thirty-four patients
(61.8%) received cryopreserved HPC products while 21
(38.2%) patients received fresh HPC products. All patients
received post-transplant cyclophosphamide on day + 3 and
day + 4, as well as tacrolimus and mycophenolate mofetil for
GVHD prophylaxis. The median duration of follow-up was
15months (range: 3–24) for the entire cohort, 14months
(range: 3–22) for the cryopreserved cohort, and 15months
(range: 8–24) for the fresh cohort. Baseline characteristics
are summarized in Table1.
There were no statistically significant differences between
the two groups in terms of age, gender, race, graft source,
ECOG score, distribution of hematologic diagnosis, disease
risk index, ABO compatibility, disease status or MRD status
prior to transplantation. Conditioning intensity and regimen
were comparable between the two groups. Among the AML
patients, there was a statistically significant difference in the
AML cytogenetics risk, with a higher proportion of patients
with favorable and adverse risk in the cryopreserved allo-
graft group, while more patients in the fresh allograft group
had intermediate risk (p < 0.03). Table2 shows graft char-
acteristics, the median CD34+, CD3+, total nucleated cell
(TNC) and percent viability of cells infused.
Hematopoietic recovery
We observed two graft failures in the recipients of cryo-
preserved grafts. One patient suffered primary graft failure
while the other experienced secondary graft failure. The two
patient’s baseline characteristics and outcomes are shown in
Table3. In patients who achieved engraftment, a statistically
significant longer time to neutrophil and platelet engraft-
ments were observed in the cryopreserved as compared
to the fresh group (p = 0.01 and p < 0.0001, respectively)
(Fig.1a and Fig.1b). In the cryopreserved group, no correla-
tion was observed between delayed engraftment and longer
liquid transit time (p = 0.42, and p = 0.14, respectively)
(Table4).
Poor graft function occurred in 44.1% of cryopreserved
allografts, while only 28.6% of fresh allografts had poor
graft function (p = 0.27). We observed a significant differ-
ence in the cumulative incidence of red blood cell (RBC)
transfusions after day + 60 between the two groups (67.6%
in the cryopreserved group vs. 28.6% in the fresh group,
p = 0.01). The difference in the cumulative incidence of
donor lymphocyte infusion (DLI) between the two groups
was also significant (p = 0.03). Of the 12 patients who
required DLI in the cryopreserved group, six (50%) were
due to poor chimerism or poor graft function, while the rest
(50%) were due to relapse. Only one patient (4.8%) in the
fresh group received DLI for relapse treatment. No differ-
ence was found in terms of cumulative incidence of second
allo-HSCT (p = 1.00). Four patients (11.8%) in the cryopre-
served allograft group underwent a second allo-HSCT, two
(50%) as a result of relapse and two (50%) due to graft fail-
ure. In the fresh allograft group, two patients (9.6%) under-
went second allo-HSCT due to relapse.
GVHD
There were no significant differences in the cumulative inci-
dence of aGVHD or cGVHD between the two groups. The
cumulative incidence of aGVHD grade II-IV was 58.8%
in the cryopreserved group and 42.9% in the fresh group
(p = 0.28). Grade III-IV aGVHD was observed in four
(11.7%) and two (9.5%) patients (p = 1.00), respectively.
The cumulative incidence of cGVHD was 64.7 and 76.2%
(p = 0.55), and the cumulative incidence of moderate to
severe cGVHD was 49.4% and 9.5% (p = 0.10) in the two
groups, respectively (Table5).
NRM
The cumulative incidence of NRM at 1-year was 12.8%
in the cryopreserved allograft group and 6.3% in the fresh
allograft group, which did not reach statistical significance
[hazard ratio (HR) = 0.94 with 95% confidence interval (CI)
0.29–34.18, p = 0.35] (Fig.2). In the cryopreserved allo-
graft group, three patients died from infection and one died
from cardiac arrest, while both patients in the fresh allograft
group died from infection. The incidence of cytomegalovirus
(CMV) reactivation by day + 100 was not significantly dif-
ferent between the two groups (p = 0.46).
M.Guo et al.
1 3
Table 1 Baseline characteristics
of patient population Characteristic Cryopreserved
n = 34
Fresh
n = 21
p-Value
Median age, years (range) 55 (21–74) 53 (19–70) 0.70
Gender, n (%) 0.16
Male 22 (64.7) 9 (42.9)
Female 12 (35.3) 12 (57.1)
Race, n (%) 1.00
White 25 (73.5) 15 (71.4)
African American 4 (11.8) 2 (9.5)
Hispanic 1 (2.9) 0 (0.0)
Other 4 (11.8) 4 (19.0)
Graft source, n (%) 0.33
MRD 4 (11.8) 1 (4.8)
MUD 18 (52.9) 8 (38.1)
MMRD 0 (0.0) 1 (4.8)
Haploidentical 12 (35.3) 11 (52.3)
ABO compatibility, n (%) 0.16
Matched 12 (35.3) 13 (61.9)
Minor mismatch 11 (32.4) 4 (19.0)
Major mismatch 11 (32.4) 4 (19.0)
Bidirectional 0 (0.0) 0 (0.0)
Disease risk index, n (%) 0.09
Low 8 (23.5) 3 (14.3)
Intermediate 9 (26.5) 12 (57.1)
High 17 (50.0) 6 (28.6)
Disease, n (%) 0.49
AML 21 (61.8) 10 (47.6)
MDS 5 (14.7) 3 (14.3)
ALL 2 (5.9) 5 (23.8)
Other 6 (17.6) 3 (14.3)
Disease status, n (%) 0.75
CR 25 (73.5) 17 (81.0)
Non-CR 9 (26.5) 4 (19.0)
Minimal residual disease prior to HSCT 0.44
No 19/25 (76.0) 15/17 (88.2)
Yes 6/25 (24.0) 2/17 (11.8)
AML cytogenetics 0.03
Favorable 8/21 (38.1) 1/10 (10.0)
Intermediate 4/21 (19.0) 5/10 (50.0)
Adverse 9/21 (42.9) 2/10 (20.0)
Unknown 0/21 (0.0) 2/10 (20.0)
ECOG, n (%) 0.52
 < 2 32 (94.1) 21 (100.0)
 ≥ 2 2 (5.9) 0 (0.0)
Conditioning intensity, n (%) 1.00
Myeloablative 27 (79.4) 17 (81.0)
RIC 7 (20.6) 4 (19.0)
Conditioning regimen 0.13
Cryopreserved versusfresh peripheral blood allogeneic stem cell transplantation outcomes…
1 3
Overall andrelapse‑free survivals
With a median follow-up of 15months for the entire cohort,
there was a statistically significant difference in OS favoring
the fresh allograft recipients (p = 0.02) (Fig.3a). The effect
of cryopreservation remained significant after multivari-
ate adjustment for age, disease status at time of transplant,
moderate to severe cGVHD and CMV infection by day 100
(HR 2.27 with 95% CI 1.23–16.51, p = 0.02). By day + 100,
94.1 and 100% of patients were alive, while at 1-year post-
transplant, 67.6% and 90.4% of patients were alive in the
cryopreserved and fresh allograft groups, respectively.
No significant difference in RFS was noted (p = 0.11,
Fig.3b; after multivariate adjustment, HR 1.61 with 95%
CI 0.84–6.07, p = 0.11). During the study period, relapse
occurred in 15 patients (44.1%) who received cryopreserved
cells with a median time to relapse of 5-months (range:
1–12), while six patients (28.6%) in the fresh group relapsed
with a median time to relapse of 7-months (range: 1–11).
By the censor date, 11 patients had died from relapse in the
cryopreserved allograft group, while two patients receiving
fresh allografts died due to relapse. Although the cumula-
tive incidence of relapse was higher in the cryopreserved
allograft group, this did not reach statistical significance
(p = 0.25, Table5).
Discussion
Cryopreservation of PBSC is not a common practice in
allogenic transplantation apart from umbilical cord blood
transplantation [11], and concerns regarding cryopreserved
products adversely affecting cell viability and effector cell
AML acute myelogenous leukemia, ALL acute lymphoblastic leukemia, CR complete remission, ECOG
eastern cooperative oncology group, MDS myelodysplastic syndrome, MMRD mismatched related donor,
MRD matched related donor, MUD matched-unrelated donor, RIC reduced intensity conditioning, Flu/Bu4
fludarabine and 4days of busulfan, Flu/Bu3 fludarabine and 3days of busulfan, Flu/Bu2 fludarabine and
2days of busulfan, Flu/TBI fludarabine and total body irradiation, Flu/Cy/TBI fludarabine, cyclophospha-
mide, and total body irradiation
Table 1 (continued) Characteristic Cryopreserved
n = 34
Fresh
n = 21
p-Value
Flu/Bu4 20 (58.8) 6 (28.6)
Flu/Bu3 4 (11.8) 3 (14.3)
Flu/Bu2 2 (5.9) 1 (4.8)
Flu/TBI 3 (8.8) 8 (38.1)
Flu/Cy/TBI 4 (11.8) 3 (14.3)
Other 1 (2.9) 0 (0.0)
Liquid transit time, hours, median (range) 22.3
(1.75–84.03)
NA NA
Time to infusion, hours, median (range) 261.86
(53.33–2447.77)
24.35
(2.57–215.13)
< 0.0001
Table 2 Graft characteristics
of fresh and cryopreserved
peripheral blood stem cell
products
NA not applicable, TNC total nucleated cell
Characteristic Cryopreserved
n = 34
Fresh
n = 21
p-Value
Pre-freeze CD34+ cell dose, × 106/kg,
median (range)
5.05 (2.71–10.19) NA NA
Infused CD34+ cell dose, × 106/kg,
median (range)
5.05 (2.11–8.56) 6.36 (5.00–12.27) 0.01
Infused CD3+ cell dose, × 108/kg,
median (range)
1.34 (0.50–3.73) 1.24 (0.51–3.49) 0.53
Infused TNC dose, × 108/kg,
median (range)
5.48 (2.28–12.66) 5.00 (2.88–12.05) 0.03
Infused TNC Viability, %,
median (range)
83 (69–94) 100 (100–100) < 0.001
M.Guo et al.
1 3
function has been the main reason for performing fresh allo-
graft infusions in these patients [14].
We retrospectively assessed the impact of cryopreserva-
tion on patient outcomes during the COVID-19 pandemic
in the year 2020. Our findings suggested that cryopreser-
vation was associated with a negative impact on allogeneic
PBSC transplant. We observed a longer time to neutro-
phil engraftment and a significantly longer time to platelet
engraftment. While some studies have suggested no differ-
ence in either neutrophil or platelet engraftment between
cryopreserved and fresh allografts [210], our findings
are supported by others who have shown outcomes con-
sistent with our observations. A large retrospective study
conducted by Hsu etal. analyzed outcomes from 1051
frozen PBSC allografts derived from related donors and
another 678 allografts derived from matched-unrelated
donors [5]. This study showed delayed platelet engraft-
ment in cryopreserved-related donor PBSC grafts and
Table 3 Summary of cases with graft failure in cryopreserved group
AML acute myelogenous leukemia, CR complete remission, Flu/Bu4 fludarabine and 4days of busulfan, Flu/Bu3 fludarabine and 3days of busulfan, Haplo haploidentical, MUD matched-unre-
lated donor, s/p status post
Gender/Age, year Disease AML cytogenetics Remission
status at trans-
plant
Condi-
tioning
regimen
Donor type CD34+ cell dose
pre-frozen, × 106/
kg
CD34+ cell dose
infused, × 106/kg
Total nucleated cell
dose infused, × 108/
kg
Graft failure Outcomes
Male/54 AML Favorable CR Flu/Bu3 Haplo 5.43 4.34 8.54 Primary Alive s/p 2nd transplant
Female/62 AML Favorable CR Flu/Bu4 MUD 10.19 8.56 3.69 Secondary Alive s/p 2nd transplant
Fig. 1 a Cumulative incidence of neutrophil engraftment; b Cumula-
tive incidence of platelet engraftment
Cryopreserved versusfresh peripheral blood allogeneic stem cell transplantation outcomes…
1 3
Table 4 Univariate linear
regression analysis of liquid
transit time with neutrophil
and platelet engraftment in
cryopreserved patients
CI confidence interval
Neutrophil engraftment Platelet engraftment
r2 Regression coefficient
(95% CI)
p-Value r2 Regression coefficient
(95% CI)
p-Value
Liquid transit time 0.02 −27.254
(−95.68–41.20)
0.42 0.07 −1.24
(-2.91–0.42)
0.14
Table 5 Transplant outcomes
RBC red blood cells, aGVHD acute graft-versus-host disease, cGVHD chronic graft-versus-host disease,
CMV cytomegalovirus
*Two patients in the cryopreserved group were excluded from time to neutrophil engraftment: one patient
who experienced primary graft failure and one patient who experienced early relapse (within one month of
transplant).
**Four patients in the cryopreserved group did not achieve platelet engraftment and were excluded from
time to platelet engraftment: one patient who experienced primary graft failure, one patient who experi-
enced secondary graft failure, and two patients who experienced relapse.
Cryopreserved
n = 34
Fresh
n = 21
p-Value
Median time to neutrophil engraftment, days (range) 21 (13–34)* 18 (15–27) 0.05
Median time to platelet engraftment, days, (range) 39.5 (18–140)** 20 (14–382) < 0.0001
Graft function, n (%)
Graft failure 2 (5.9) 0 (0.0) 0.52
Poor graft function 15 (44.1) 6 (28.6) 0.27
Donor lymphocyte infusion, n (%) 0.03
Yes, for relapse 7 (20.6) 1 (4.8)
Yes, for poor chimerism 5 (14.7) 0 (0.0)
No 22 (64.7) 20 (95.2)
RBC transfusion after day + 60, n (%) 0.01
Yes 23 (67.6) 6 (28.6)
No 11 (32.4) 15 (71.4)
aGVHD grade II-IV, n (%) 0.28
No 20 (58.8) 9 (42.9)
Yes 14 (41.2) 12 (57.1)
aGVHD grade III-IV, n (%) 4 (11.7) 2 (9.5) 1.00
cGVHD, total, n (%) 0.55
No 22 (64.7) 16 (76.2)
Yes 12 (35.3) 5 (23.8)
cGVHD, moderate to severe, n (%) 10 (49.4) 2 (9.5) 0.10
CMV reactivation by day + 100, n (%) 0.46
No 27 (79.4) 19 (90.5)
Yes 7 (20.6) 2 (9.5)
Second stem cell transplant, n (%) 1.00
Yes 4 (11.8) 2 (9.6)
No 30 (88.2) 19 (90.5)
Relapse, n (%) 0.25
Yes 15 (44.1) 6 (28.6)
No 19 (55.9) 15 (71.4)
Death, n (%) 0.06
Yes 15 (44.1) 4 (19.0)
No 19 (55.9) 17 (81.0)
M.Guo et al.
1 3
delayed engraftment of both neutrophils and platelets in
cryopreserved unrelated donor PBSC grafts. Dagdas etal.
also showed longer neutrophil engraftment kinetics using
frozen products [11].
Two graft failures were observed in the cryopreserved
group while none were observed in the fresh group. Impor-
tantly, poor graft function occurred in 44.1% of cryopre-
served allografts as compared to 28.6% of fresh allografts.
Twice as many cryopreserved recipients required prolonged
RBC transfusions after day + 60 (67.6 vs. 28.6%, respec-
tively). Additionally, more cryopreserved recipients required
DLI than recipients of fresh stem cells. Consistent with our
data, Lioznov etal. observed more graft failures in cryo-
preserved allograft recipients compared to fresh allograft
recipients (27 vs 1.4%, respectively) [13].
With a median follow-up of 15months, a significant
decrease in OS was noted in the cryopreserved group com-
pared with the fresh group, while NRM and RFS did not
reach statistical significance. Hsu etal. found minimal
impact of cryopreservation in related PBSC allografts in
terms of NRM, relapse, OS and RFS, but noted increased
relapse as well as decreased RFS and OS in cryopreserved
unrelated PBSC allografts [5]. The authors suspected a
potentially longer transit time of unrelated donor grafts prior
to cryopreservation, the decline in post-thaw colony forming
units, and other patient-related reasons for cryopreservation
may have played a role in the worse outcomes noted in their
study [5]. In our study, although the cryopreserved cells had
a slightly lower number of CD34+ cell infused, as expected
due to the freeze-thawing process, this cannot explain our
findings. Prior data have shown that the minimum acceptable
dose of CD34+ cells for successful transplantation is above
2.0 × 106 cells/kg [19]. Even though all of our post-thawed
products met this criteria, the transplant outcomes differed
significantly between the cryopreserved and fresh groups,
suggesting in cryopreserved allografts, other factors besides
CD34+ cell dose may play a role. The cryo-protective agent
used in the cryopreservation process could result in adverse
reactions and potentially have a negative impact on survival
outcomes [20]. It is important to note that all patients in
our study received post-transplant cyclophosphamide as part
of GVHD prophylaxis. As mentioned above, Linzov etal.,
demonstrated that graft failures in the cryopreserved cohort
appeared to occur more commonly in patients who received
allografts with low aldehyde dehydrogenase bright CD34+
cells [13]. Given the effect of aldehyde dehydrogenase on
the metabolism of cyclophosphamide, it is plausible that a
combination of cryopreservation and post-transplant cyclo-
phosphamide may adversely affect engraftment. Further
investigation is needed to test this hypothesis.
Published data for the incidence of aGVHD in cryopre-
served allo-HSCT are conflicting. Parody etal. reported a
higher incidence and earlier onset of aGVHD in patients
receiving cryopreserved PBSC while the difference in the
incidence of cGVHD was not significant [12]. Medd etal.
reported comparable incidence of aGVHD but a non-sig-
nificant increase in extensive cGVHD in the cryopreserved
Fig. 2 Non-relapse mortality (NRM)
Fig. 3 a Overall survival; b Relapse-free survival
Cryopreserved versusfresh peripheral blood allogeneic stem cell transplantation outcomes…
1 3
PBSC group [14]. Alotaibi etal. reported no difference in
grade II-IV aGVHD between cryopreserved versus fresh
allografts, but did note a higher incidence of moderate/
severe cGVHD in the cryopreserved group [2]. Our study
demonstrated no statistically significant differences in grade
II-IV aGVHD or cGVHD.
While our study population was relatively homogenous
and the rational for cryopreservation was predominantly due
to the impact of COVID-19, as opposed to heterogenous rea-
sons requiring cryopreservation, there are several limitations
to our work. Our study is limited by its retrospective nature,
small sample size, and relatively short duration of follow-
up. Additionally, the use of post-transplant cyclophospha-
mide prevents us from generalizing our conclusions to other
GVHD prophylaxis regimens.
In summary, our study showed decreased OS, delayed
engraftment, and a higher need for DLI and RBC trans-
fusions in patients undergoing cryopreserved PBSC allo-
HSCT with post-transplant cyclophosphamide. We suggest
cryopreserved allogeneic transplantation to be performed
with caution. Further prospective studies are required to fur-
ther investigate the impact of cryopreservation in allogeneic
PBSC transplantation.
Acknowledgements This research did not receive any specific grant
from funding agencies in the public, commercial, or not-for-profit
sectors.
Author contributions MG: data collection, methodology, investigation,
writing, review. JL: data collection, methodology, investigation, writ-
ing, review. PC: data collection, methodology, investigation, writing,
review. SA: writing, review. RDP: writing, review. SM: conceptualiza-
tion, methodology, supervision, writing, review and editing.
Data availability statement The data us accurate and available upon
request.
Declarations
Conflict of interest The authors declare that they have no conflict of
interest.
References
1. Wiercinska E, Schlipfenbacher V, Bug G, Bader P, Verbeek M,
Seifried E, etal. Allogeneic transplant procurement in the times of
COVID-19: quality report from the central European cryopreser-
vation site. J Transl Med. 2021;19:145. https:// doi. org/ 10. 1186/
s12967- 021- 02810-9.
2. Alotaibi AS, Prem S, Chen S, Lipton JH, Kim DD, Viswa-
bandya A, etal. Fresh vs. frozen allogeneic peripheral blood
stem cell grafts: A successful timely option. Am J Hematol.
2021;96:179–87.
3. Kim DH, Jamal N, Saragosa R, Loach D, Wright J, Gupta V,
etal. Similar outcomes of cryopreserved allogeneic peripheral
stem cell transplants (PBSCT) compared to fresh allografts. Biol
Blood Marrow Transplant. 2007;13:1233–43.
4. Devine SM. Transplantation of allogeneic cryopreserved
hematopoietic cell grafts during the COVID-19 pandemic: a
National Marrow Donor Program perspective. Am J Hematol.
2021;96:169–71.
5. Hsu JW, Farhadfar N, Murthy H, Logan BR, Bo-Subait S, Frey
N, etal. The effect of donor graft cryopreservation on alloge-
neic hematopoietic cell transplantation outcomes: a center for
international blood and marrow transplant research analysis.
Implications during the COVID-19 pandemic. Transplant Cell
Ther. 2021;27:507–16.
6. Hamadani M, Zhang MJ, Tang XY, Fei M, Brunstein C, Chhabra
S, etal. Graft cryopreservation does not impact overall survival
after allogeneic hematopoietic cell transplantation using post-
transplantation cyclophosphamide for graft-versus-host disease
prophylaxis. Biol Blood Marrow Transplant. 2020;26:1312–7.
7. Valentini CG, Chiusolo P, Bianchi M, Metafuni E, Orlando N,
Giammarco S, etal. Coronavirus disease 2019 pandemic and
allogeneic hematopoietic stem cell transplantation: A single
center reappraisal. Cytotherapy. 2021;23:635–40.
8. Jacob RP, Flynn J, Devlin SM, Maloy M, Giralt SA, Maslak
P, etal. Universal engraftment after allogeneic hematopoietic
cell transplantation using cryopreserved CD34-selected grafts.
Transplant Cell Ther. 2021;27:697-e1.
9. Fernandez-Sojo J, Azqueta C, Valdivia E, Martorell L, Medina-
Boronat L, Martínez-Llonch N, etal. Cryopreservation of
unrelated donor hematopoietic stem cells: The right answer for
transplantations during the COVID-19 pandemic? Bone Marrow
Transplant. 2021;56:2489–96.
10. Eapen M, Zhang MJ, Tang XY, Lee SJ, Fei MW, Wang HL,
etal. Hematopoietic cell transplantation with cryopreserved
grafts for severe aplastic anemia. Biol Blood Marrow Trans-
plant. 2020;26:e161-6.
11. Dagdas S, Ucar MA, Ceran F, Gunes AK, Falay M, Ozet G.
Comparison of allogenic stem cell transplantations performed
with frozen or fresh stem cell products with regard to GVHD
and mortality. Transfus Apher Sci. 2020;59: 102742. https:// doi.
org/ 10. 1016/j. trans ci. 2020. 102742.
12. Parody R, Caballero D, Márquez-Malaver FJ, Vázquez L,
Saldaña R, Madrigal MD, etal. To freeze or not to freeze
peripheral blood stem cells prior to allogeneic transplantation
from matched related donors. Eur J Haematol. 2013;91:448–55.
13. Lioznov M, Dellbrügger C, Sputtek A, Fehse B, Kröger N,
Zander AR. Transportation and cryopreservation may impair
haematopoietic stem cell function and engraftment of allogeneic
PBSCs, but not BM. Bone Marrow Transplant. 2008;42:121–8.
14. Medd P, Nagra S, Hollyman D, Craddock C, Malladi R.
Cryopreservation of allogeneic PBSC from related and unre-
lated donors is associated with delayed platelet engraftment
but has no impact on survival. Bone Marrow Transplant.
2013;48:243–8.
15. National Marrow Donor Program (NMDP). Response to COVID-
19: Up-to- date information for all Network partners on coronavi-
rus impacts. Available at: https:// netw o r k. bethe match clini cal. org/
news/ nmdp/ be- the- match- respo nse- to- covid- 19/. Accessed Oct 1,
2020
16. Valcárcel D, Sureda A. Graft failure. In: Carreras E, Dufour C,
Mohty M, Kröger N, editors. The EBMT handbook hematopoietic
stem cell transplantation and cellular therapies. 7th ed. Cham:
Springer; 2019. p. 307–13.
17. Przepiorka D, Weisdorf D, Martin P, Klingemann HG, Beatty P,
Hows J, Thomas ED. 1994 consensus conference on acute GVHD
grading. Bone Marrow Transplant. 1995;15(6):825–8.
18. Alessandrino EP, Bernasconi P, Caldera D, Colombo A, Bonfichi
M, Malcovati L, etal. Adverse events occurring during bone mar-
row or peripheral blood progenitor cell infusion: analysis of 126
cases. Bone Marrow Transplant. 1999;23:533–7.
M.Guo et al.
1 3
19. Singhal S, Powles R, Treleaven J, Kulkarni S, Sirohi B, Horton
C, etal. A low CD34+ cell dose results in higher mortality and
poorer survival after blood or marrow stem cell transplantation
from HLA-identical siblings: should 2×106 CD34+ cells/kg be
considered the minimum threshold? Bone Marrow Transplant.
2000;26:489–96.
20. Shu Z, Heimfeld S, Gao D. Hematopoietic SCT with cryopre-
served grafts: Adverse reactions after transplantation and cryo-
protectant removal before infusion. Bone Marrow Transplant.
2014;49:469–76.
Publisher's Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
Springer Nature or its licensor (e.g. a society or other partner) holds
exclusive rights to this article under a publishing agreement with the
author(s) or other rightsholder(s); author self-archiving of the accepted
manuscript version of this article is solely governed by the terms of
such publishing agreement and applicable law.
... In Figure 2, we summarized the findings of all studies included in our revision regarding neutrophil and platelet engraftment. The effects of cryopreservation on acute and chronic GVHD and survival and relapse rates are recapitulated in Tables 1 and 2. [86,88,89,[91][92][93][94][95][96]98,99,101,102,[104][105][106][107][108][109][110][111]. Data are given as median and range, median and 95% confidence interval a , or median and interquartile range b . ...
... The study of Maurer (2023) [95] reported no data on platelet engraftment. [86,88,89,[91][92][93][94][95][96]98,99,101,102,[104][105][106][107][108][109][110][111]. Data are given as median and range, median and 95% confidence interval a , or median and interquartile range b . ...
... Therefore, the authors concluded that the inferior outcomes of cryopreserved transplants were possibly due to the combination of ATG and PTCy impacting the differential tolerance to cryopreservation of various components of the grafts [100]. Similar findings were reported by Guo et al. in a single center study involving 34 patients receiving cryopreserved PBSC transplant and 31 controls [101]. Conversely, various single-institution studies found no differences between cryo-and fresh grafts in unrelated donor transplants [102][103][104][105][106]. A further single institution study reported delayed engraftment and a trend toward poorer immuno-reconstitution of cryopreserved allografts overall in myeloablative conditioning transplant [107]. ...
Article
Full-text available
The COVID-19 pandemic has precipitously changed the practice of transplanting fresh allografts. The safety measures adopted during the pandemic prompted the near-universal graft cryopreservation. However, the influence of cryopreserving allogeneic grafts on long-term transplant outcomes has emerged only in the most recent literature. In this review, the basic principles of cell cryopreservation are revised and the effects of cryopreservation on the different graft components are carefully reexamined. Finally, a literature revision on studies comparing transplant outcomes in patients receiving cryopreserved and fresh grafts is illustrated.
Article
Full-text available
The COVID-19 pandemic has had a significant impact on medical practices, including the delivery of allogeneic hematopoietic cell transplantation (HCT). In response, transplant centers have made changes to their procedures, including an increased use of cryopreservation for allogeneic haematopoietic progenitor cell (HPC) grafts. The use of cryopreserved grafts for allogeneic HCT has been reviewed and analysed in terms of potential benefits and drawbacks based on existing data on impact on cell subsets, hematological recovery, and clinical outcomes of approximately 2000 patients from different studies. A survey of European Society for Blood and Marrow Transplantation centers was also conducted to assess changes in practice during the pandemic and any unnecessary burdens on HPC donors. Before the pandemic, only 7.4% of transplant centers were routinely cryopreserving HPC products, but this percentage increased to 90% during the pandemic. The results of this review and survey suggest that cryopreservation of HPC grafts is a viable option for allogeneic HCT in certain situations, but further research is needed to determine long-term effects and ethical discussions are required to balance the needs of donors and patients when using frozen allografts.
Article
Full-text available
Cryopreservation was recommended to ensure continuity of unrelated donor (UD) hematopoietic stem cell transplantation (HSCT) during COVID-19 pandemic. However, its impact on clinical outcomes and feasibility was not well known. We compared 32 patients who underwent UD HSCT using cryopreserved peripheral blood stem cells (PBSC) during the COVID-19 pandemic with 32 patients who underwent UD HSCT using fresh PBSC in the previous period. Median neutrophil engraftment was 17.5 and 17.0 days with cryopreserved and fresh grafts, respectively. Non-significant delays were found in platelet recovery days (25.5 versus 19.0; P = 0.192) and full donor chimerism days (35.0 and 31.5; P = 0.872) using cryopreserved PBSC. The rate of acute graft-versus-host disease at 100 days was 41% (95% CI [21–55%]) in cryopreserved group versus 31% (95% CI [13–46%]) in fresh group (P = 0.380). One-hundred days progression-relapse free survival and overall survival did not differ significantly. During COVID-19 pandemic, six frozen UD donations were not transfused and logistical and clinical issues regarding cryopreservation procedure, packaging, and transporting appeared. In summary, UD HSCT with cryopreserved PBSC was safe during this challenging time. More efforts are needed to ensure that all frozen grafts are transplanted and cryopreservation requirements are harmonized.
Article
Full-text available
Background Because of limitations of transportation imposed by the COVID-19 pandemic, current recommendation calls for cryopreservation of allogeneic stem cell transplants before patient conditioning. A single cell therapy laboratory was selected to function as the central cryopreservation hub for all European registry donor transplants intended for the Australian-Pacific region. We examined properties of these transplants to ascertain how quality is maintained. Methods We analyzed 100 pandemic-related allogeneic mobilized blood-derived stem cell apheresis products generated at 30 collection sites throughout Europe, shipped to and cryopreserved at our center between April and November of 2020. Products were shipped in the cool, subsequently frozen with DMSO as cryoprotectant. Irrespective of origin, all products were frozen within the prescribed shelf-life of 72 h. Results Prior to cryopreservation, viable stem cell and leukocyte count according to the collection site and our reference laboratory were highly concordant (r ² = 0.96 and 0.93, respectively) and viability was > 90% in all instances. Median nominal post-thaw recovery of viable CD34+ cells was 42%. Weakly associated with poorer CD34+ cell recovery was higher leukocyte concentration, but not time lag between apheresis or addition of cryopreservant, respectively, and start of freezing. The correlation between pre- and post-thaw CD34+ cell dose was high (r ² = 0.85), hence predictable. Neutrophil and platelet engraftment were prompt with no evidence of dose dependency within the range of administered cell doses (1.31–15.56 × 10 ⁶ CD34+ cells/kg). Conclusions General cryopreservation of allogeneic stem cell transplants is feasible. While more than half of the CD34+ cell content is lost, the remaining stem cells ensure timely engraftment.
Article
Full-text available
Cryopreservation of grafts has been established in autologous and cord blood transplantation, yet there is little experience regarding the effect of cryopreservation with sibling and unrelated grafts. We evaluated the effect of cryopreservation of grafts on allogeneic transplant outcomes using related, unrelated and haploidentical donors, including 958 patients, age 18‐74 years (median 55) and using PBSC for various hematologic malignancies. Fresh grafts were received by 648 (68%) patients, 310 (32%) received cryopreserved. There was no difference between fresh vs cryopreserved grafts for neutrophil engraftment (P = .09), platelet engraftment (P = .11), graft failure (5.6% vs 6.8%, P = .46) and grade II‐IV acute graft‐vs‐host disease (GVHD) (P = .71), moderate/severe chronic GVHD was observed in 176 (27%) vs 123 (40%) patients, respectively (P < .001). Multivariable analysis demonstrated no difference between fresh vs cryopreserved for OS (P = .39) and CIR (P = .08) while fresh grafts demonstrated borderline increased NRM (HR 1.27, 95% CI 1.02‐1.59, P = .04). Of note, for patients with no or mild chronic GVHD, CIR was less for fresh compared to cryopreserved (HR = 0.67 for fresh, 95% CI 0.48‐0.92, P = .01). We conclude there were no differences in engraftment and survival between fresh and cryopreserved grafts for allogeneic HCT, thus establishing cryopreservation to be a safe option for allogeneic HCT.
Article
Full-text available
The COVID-19 pandemic and the ensuing barriers to the collection and transport of donor cells, it is often necessary to collect and cryopreserve grafts before initiation of transplant conditioning. The effect on transplant outcomes in non-malignant disease is unknown. This analysis examined the effect of cryopreservation of related and unrelated donor grafts for transplantation for severe aplastic anemia in the US during 2013-2019. Included are 52 recipients of cryopreserved grafts who were matched for age, donor type, and graft type to 194 recipients who received non-cryopreserved grafts. Marginal Cox regression models were built to study the effect of cryopreservation and other risk factors associated with outcomes. We recorded higher 1-year rates of graft failure (HR 2.26, 95% CI 1.17 – 4.35, p=0.01) and of 1-year overall mortality (HR 3.13, 95% CI 1.60 – 6.11, p=0.0008) after transplantation of cryopreserved compared to non-cryopreserved grafts with adjustment for sex, performance score, comorbidity, cytomegalovirus serostatus, and ABO blood group match). Acute and chronic GVHD did not differ between groups. Adjusted probabilities of 1-year survival were 73% (95% 60 – 84) and 91% (95% CI 86 – 94) with cryopreserved and non-cryopreserved grafts, respectively. These data support the use of non-cryopreserved grafts, when possible, for severe aplastic anemia.
Article
Full-text available
INTRODUCTION The COVID-19 pandemic has created significant barriers to timely donor evaluation, cell collection and graft transport for allogeneic hematopoietic stem cell transplantation (allo-HCT). To ensure availability of donor cells on the scheduled date of infusion, many sites now collect cryopreserve grafts before the start of pretransplant conditioning. Post-transplant cyclophosphamide (ptCY), is an increasingly used approach for graft-versus-host disease (GVHD) prophylaxis but the impact of graft cryopreservation on the outcomes of allo-HCT using ptCY is not known. Using the Center for International Blood and Marrow Transplant Research (CIBMTR) database, we compared the outcomes of HCT using cryopreserved versus fresh grafts in patients undergoing HCT for hematologic malignancy with ptCY. METHODS We analyzed 274 patients with hematologic malignancy undergoing allo-HCT from 2013-2018 with cryopreserved grafts and ptCY. Eighteen received bone marrow and 256, peripheral blood grafts. These were matched for age, graft type, disease risk index (DRI) and propensity score to 1,080 patients that underwent allo-HCT with a fresh graft. The propensity score, which is an assessment of the likelihood of receiving a fresh versus cryopreserved graft, was calculated logistic regression to account for the following: disease histology, Karnofsky Performance Score (KPS), HCT-comorbidity index, conditioning regimen intensity, donor type and recipient race. The primary endpoint was overall survival (OS). Secondary endpoints included acute and chronic graft-versus-host disease GVHD, non-relapse mortality (NRM), relapse/progression and disease-free survival (DFS). Because of multiple comparisons, only p-values <0.01 were considered statistically significant. RESULTS The two cohorts (cryopreserved versus fresh) were similar in patient age, KPS, diagnosis, DRI, HCT-comorbidity index, donor/graft source, and conditioning intensity. One-year probabilities of OS were 71.1% (95% confidence interval, 68.3-73.8%) with fresh grafts and 70.3% (64.6-75.7%) with cryopreserved grafts (p=0.81). Corresponding probabilities of OS survival at two years were 60.6% (57.3-63.8%) and 58.7% (51.9-65.4%) (p=0.62). In matched pair regression analysis, graft cryopreservation was not associated with a significantly higher risk of mortality (Hazard Ratio for cryopreserved versus fresh [HR] =1.05, 95% confidence interval, 0.86-1.29, p=0.60). Similarly, rates neutrophil recovery (HR=0.91, 0.80-1.02, p=0.12), platelet recovery (HR=0.88, 0.78-1.00, p=0.05), grade 3-4 acute GVHD (HR=0.78, 0.50-1.22, p=0.27), NRM (HR=1.16, 0.86-1.55, p=0.32) and relapse/progression (HR=1.21, 0.97-1.50, p=0.09) were similar with cryopreserved versus fresh grafts. There were somewhat lower rates chronic GVHD (HR=0.78, 0.61-0.99, p=0.04) and DFS (HR for treatment failure=1.19, 95%CI=1.01-1.29, p=0.04) with graft cryopreservation that were of marginal statistical significance after adjusting for multiple comparisons. CONCLUSIONS Graft cryopreservation does not significantly delay hematopoietic recovery, increase acute GVHD risk or NRM, or decrease overall survival after all-HCT using ptCY.
Article
Background As a result of the COVID-19 pandemic, most centers performing allogeneic hematopoietic cell transplant (allo-HCT) have switched to the use of cryopreserved grafts. Previous investigators have suggested that cryopreserved allografts may heighten risk of nonengraftment. To date, no study has investigated the effect of cryopreservation of CD34-selected hematopoietic progenitor cells (CD34+HPC) used as the sole graft source. Objective We sought to evaluate outcomes after unrelated donor or matched sibling allo-HCT with cryopreserved CD34+HPC. Study Design This was a single center analysis of adult patients with hematologic malignancies who underwent allo-HCT with cryopreserved CD34 selected allo-HCT grafts from 1/2010 to 6/2017. All patients received ablative conditioning and antirejection prophylaxis with rabbit ATG. GCSF-mobilized leukapheresis products underwent CD34 selection using the CliniMACS Reagent System. Cells were then cryopreserved in DMSO (final concentration 7.5%) to -90°C using a controlled rate freezing system before transfer to vapor phase liquid nitrogen storage. In internal validation, this method has shown 92% mean CD34+ viability and 99.7% mean CD34+ recovery. Engraftment was defined as the first of 3 consecutive days of ANC ≥ 0.5. Platelet recovery was recorded as the first of 7 consecutive days with platelet count ≥ 20 K/mcL without transfusion. Kaplan-Meier methodology was used to estimate OS/RFS, and cumulative incidence functions were used to estimate relapse, NRM, and acute GvHD. Results A total of 64 patients received a cryopreserved CD34 selected graft. Median CD34+ cell count before cryopreservation was 6.6 × 10^6/kg (range 1.4–16.1) and median CD3+ count 2.0 × 10^3/kg (range 0–21.1). All patients were engrafted, at a median of 11 days post-HCT (range 8–14). One patient had poor graft function in the setting of CMV viremia, requiring CD34 selected boost on day 57. Median time to platelet recovery was 16 days (range 13–99). Comparing cryopreserved versus fresh grafts, estimated 2-year OS was 70% (58-83%, 95% CI) vs 62% (57-67%, 95% CI) with fresh grafts (HR 0.86, 0.54-1.35 95% CI, p = 0.5). The estimated 2-year RFS was 59% (48-74%, 95% CI) versus 56% (51-61%, 95% CI; HR 1.01, 0.68-1.51 95% CI, p > 0.9). Cumulative incidence of relapse at 2 years was 29% (17-41%, 95% CI) versus 23% (19-27%, 95% CI; p = 0.16), while cumulative incidence of NRM at 2 years was 17% (9-28%, 95% CI) versus 23% (19-28%, 95% CI; p = 0.24). Grade II-IV acute GvHD by day 100 cumulative incidence was 16% in cryopreserved (8-26%, 95% CI), and 16% (13-20%, 95% CI; p = 0.97) after fresh graft infusion. Moderate to severe chronic GvHD by day 365 occurred in only one recipient of a cryopreserved graft (2%). Conclusions In patients with hematologic malignancies who received cryopreserved allogeneic CD34+HPC, engraftment, GvHD, and survival outcomes were consistent with that seen in fresh allogeneic CD34+HPC grafts at our center. Our laboratory validation and clinical experience demonstrate the safety of our cryopreservation procedure for CD34 selected allografts.
Article
The COVID-19 pandemic has resulted in the increased use of cryopreserved grafts for allogeneic hematopoietic cell transplantation (HCT). However, information about the effect of cryopreservation on outcomes for patients receiving allogeneic donor grafts is limited. We evaluated outcomes of HCT recipients who received either fresh or cryopreserved allogeneic bone marrow or peripheral blood stem cell (PBSC) grafts reported to the CIBMTR. A total of 7,397 patients were included in the analysis. Recipients of cryopreserved graft were divided into three cohorts based on graft source: HLA matched related PBSC (n=1,051), matched unrelated PBSC (n=678), and matched related or unrelated bone marrow donors (n=154). These patients were propensity score matched with 5,514 patients who received fresh allografts. The primary endpoint was engraftment. Multivariate analyses showed no significant increased risk of delayed engraftment, relapse, NRM, or survival with cryopreservation of marrow grafts. In contrast, cryopreservation of related donor PBSC grafts was associated with decreased platelet recovery (HR=0.73, CI=0.68-0.78, p<0.001) and an increased risk of grade II-IV (HR-1.27, CI=1.09-1.48, p=0.002) and grade III-IV (HR=1.48, CI=1.19-1.84, p<0.001) acute GVHD. Cryopreservation of unrelated PBSC grafts was associated with delayed engraftment of neutrophils (HR=0.77, CI=0.71-0.84, p<0.001) and platelets (HR=0.61, CI=0.56-0.66, p<0.001) as well as an increased risk of NRM (HR=1.4, CI=1.18-1.66, p<0.001) and relapse (HR=1.32, CI=1.11-1.58, p=0.002) and decreased PFS (HR=1.36, CI=1.20-1.55, p<0.001) and OS (HR=1.38, CI=1.22-1.58, p<0.001). Reasons for cryopreservation were not routinely collected, however in a subset of unrelated donor HCT the reason was typically a change in patient condition. Products cryopreserved for patient reasons were significantly associated with inferior OS in MVA (HR=0.65, CI=0.44-0.96, P=0.029). We conclude that cryopreservation is associated with slower engraftment of PBSC grafts which may be associated with inferior transplant outcomes in some patient populations. However the small numbers in the cryopreserved BM cohort and the lack of information on the reason for cryopreservation in all patients suggests that these data should be interpreted with caution, particularly in the context of the risks associated with unexpected loss of a graft during the pandemic. Future analyses addressing outcomes when cryopreservation is universally applied are urgently required.
Article
Background: The coronavirus disease 2019 (COVID-19) pandemic has deeply modified the complex logistical process underlying allogeneic hematopoietic stem cell transplant practices. Aim: In light of these changes, the authors compared data relative to allogeneic transplants carried out from 2018 at their center before (n = 167) and during the pandemic (n = 45). Methods: The authors examined patient characteristics, donor and graft types, cell doses and main transplant outcomes. Moreover, the authors evaluated the rise of costs attributable to additional COVID-19-related procedures as well as the risk of adverse events these procedures conveyed to grafts or recipients. Results: Overall, the number of transplants did not decrease during the pandemic, whereas patients at high relapse risk were prioritized. Transplants were mainly from matched unrelated donors, with a significant decrease in haploidentical related donors. Moreover, the use of bone marrow as a graft for haploidentical transplant was almost abandoned. Cryopreservation was introduced for all related and unrelated apheresis products, with a median storage time of 20 days. Notably, transplant outcomes (engraftment, acute graft-versus-host disease and non-relapse mortality) with cryopreserved products were comparable to those with fresh products. Conclusions: Considering that the emergency situation may persist for months, cryopreserving allogeneic grafts can offer a lifesaving opportunity for patients whose allogeneic transplant cannot be postponed until after the end of the COVID-19 pandemic.
Article
Purpose Stem cells are collected from donors and infused to the recipient in allogenic peripheral stem cell transplantations. The use of frozen stem cells can promote donor compatibility, and overcoming possible problems due to insufficient stem cell mobilization will also be easier. Nevertheless, studies about the use of frozen peripheral stem cells in allogenic transplantation are extremely rare. In this study, we aimed to compare the clinical outcomes of allogenic stem cell transplants from frozen or fresh stem cell products. Materials and Methods This retrospective analysis was conducted between April 2004 and September 2018 in the bone marrow transplantation unit of Ankara Numune Training and Research Hospital. Clinical data of patients who received allogenic peripheral stem cell transplantations from fully matched sibling donors were compared for 42 fresh and 30 frozen stem cell transplants. Results While the platelet engraftment period, febrile neutropenia period, hospitalization period, and 100-day mortality rates did not show any differences, the neutrophil engraftment period was longer in the frozen group (mean: 14 days vs. 16 days, p = 0.006). Acute and chronic graftversus-host disease (GVHD) rates were similar in both groups; however, the rate of grade 3 or4 chronic liver GVHD was slightly higher in transplants performed with fresh stem cells compared to the frozen group (p = 0.046). Overall survival was similar between the groups (p = 0.700). Conclusion The use of frozen peripheral stem cells in allogenic stem cell transplantation may be a reasonable option that can be applied without causing a significant change in clinical results.