Pi is 2666636722016669

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Transplantation and Cellular Therapy 29 (2023) 46.e1 46.

e6

Transplantation and
Cellular Therapy
journal homepage: www.tctjournal.org

Full Length Article


Pediatric

Durable Engraftment and Excellent Overall Survival After CD34-


Selected Peripheral Blood Stem Cell Boost in Pediatric Patients With
Poor Graft Function Following Allogeneic Stem Cell Transplantation
Ellen Fraint1,2, Sana Farooki1,3, Elizabeth Klein1, Audrey Mauguen4, Susan E Prockop1,5,6,
Andromachi Scaradavou1,5, Kevin Curran1,5, Maria Cancio1,5, Barbara Spitzer1,5, Jaap Jan Boelens1,5,
Joseph Oved1,5, Andrew Harris1,5, Richard J O’Reilly1,5, Nancy A. Kernan1,5,*
1
Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service, New York, New York
2
Division of Pediatric Hematology, Oncology, and Cellular Therapy, Children’s Hospital at Montefiore and Albert Einstein College of Medicine, Bronx New York
3
Division of Pediatric Hematology/Oncology, Charleston Area Medical Center, West Virginia University, Charleston, West Virginia
4
Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
5
Department of Pediatrics, Weill Cornell Medicine, New York, New York
6
Dana Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts

Article history: A B S T R A C T
Received 3 August 2022 Poor graft function (PGF) is a life-threatening complication after allogeneic stem cell transplantation (alloSCT).
Accepted 28 September 2022 Historically, outcomes of patients with PGF have been very poor, and there are no standardized approaches to
treatment. Furthermore, few outcomes after CD34-selected stem cell boost (CD34+SCB) for PGF in pediatric
Key Words: alloSCT recipients have been reported. Here we report on a single center experience with CD34+SCB for PGF after
Pediatric allogeneic alloSCT in patients treated on the Pediatric Transplant and Cellular Therapy Service at MSK Kids, Memorial Sloan
transplantation Kettering Cancer Center. A retrospective analysis of patients transplanted for malignant and nonmalignant disor-
Poor graft function (PGF) ders who received a CD34+SCB between 2008 to 2020 for treatment of PGF defined as the need for granulocyte
CD34-selected peripheral blood
colony-stimulating factor (G-CSF) and/or packed red blood cell or platelet transfusion support with bone marrow
stem cell boosts
donor chimerism 85%. Peripheral blood stem cells from the original donor were the source for CD34+SCB. Dura-
ble complete recovery (durable CR) was defined as recovery of peripheral blood counts without recurrent need
for G-CSF or transfusion support. The main outcomes of interest were recovery of hematopoiesis and overall sur-
vival. Development of graft versus host disease (GVHD) was an additional outcome of interest. Fourteen patients
with PGF received a boost. Six patients had no known infection, while 8 patients had PGF associated with an infec-
tion. The probability of CR at 60 days was 79% (95% confidence interval [CI], 57%-100%). The overall survival at
both 2 and 5 years was 78% (95% CI, 56%-100%). One patient developed GVHD, which was fatal. No other CD34
+SCB-related toxicities were observed. While including patients with PGF as recently defined by the American
Society for Transplantation and Cellular Therapy, as well as PGF in patients with concomitant infections, we dem-
onstrate that CD34+SCB is safe and can provide for durable trilineage hematopoietic recovery and long-term sur-
vival in pediatric patients after alloSCT.
© 2022 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights
reserved.

Poor graft function (PGF) is a life-threatening complication multiple transfusions [1,2]. Studies have defined PGF as either
after allogeneic stem cell transplantation (alloSCT) character- cytopenia of at least 2 cell types (absolute neutrophil count
ized by cytopenias, transfusion dependence, and hypocellular [ANC]  500 £ 106/L; platelet count  20 £ 109/L; or hemoglo-
marrow, which can be associated with the development of bin (HB)  7 g/dL) or the need for transfusion support beyond
severe infections, hemorrhage, and iron overload secondary to day 28 after SCT in recipients of bone marrow (BM) or periph-
eral blood stem cells or day 42 after SCT in recipients of umbili-
cal cord blood [2 6]. Recently, a standardized definition for
Financial disclosure: See Acknowledgments on page 46.e6. PGF was established by the American Society for Transplanta-
*Correspondence and reprint requests: Nancy A. Kernan, MSK Kids, Memo- tion and Cellular Therapy (ASTCT) as frequent dependence on
rial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065.
E-mail address: kernann@mskcc.org (N.A. Kernan).
blood or platelet transfusions or growth factor support with

https://doi.org/10.1016/j.jtct.2022.09.027
2666-6367/© 2022 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.
E. Fraint et al. / Transplantation and Cellular Therapy 29 (2023) 46.e1 46.e6 46.e2

donor myeloid and lymphoid chimerism within a desirable The main outcomes of interest were the achievement of CR, incidence of
target level and in the absence of other explanations, such as GVHD, and overall survival. The cumulative incidence of CR was estimated
using an Aalen-Johansen estimator (with death and second alloSCT as com-
disease relapse, drugs, or infections [4]. Early PGF is defined as
peting events). Overall survival (OS) was calculated as the time from CD34
slow or incomplete recovery of BM function beyond day 28, +SCB to death from any cause, and surviving patients were censored at their
whereas late PGF is defined as a loss of BM function after ini- date of last contact. Survival rates were estimated using a Kaplan-Meier esti-
tially achieving normal BM function. There are no standardized mator. The median follow-up time was calculated by reverse Kaplan-Meier.
Patients alive without recovery were censored at their date of last contact.
curative approaches for patients with either type of PGF, and
supportive care with growth factors and transfusions is a
mainstay of treatment. Repeat alloSCT after preparative condi- RESULTS
tioning can successfully improve PGF with response rates of Fourteen consecutive patients who underwent alloSCT and
66% to 100%, but a high rate of GVHD and infectious complica- subsequently developed PGF received treatment with CD34
tions lead to high non-relapse mortality (NRM) and poor over- +SCBs for PGF (Tables 1-3). Of note, 6 patients had PGF as
all survival (20%-25%) [7 9]. defined by the ASTCT without infection and 8 patients had PGF
Infusion of an additional dose (boost) of unmodified
hematopoietic stem cells (SCB) or CD34-selected peripheral Table 1
blood stem cells (CD34+SCB) from the original donor, often Summary Patient Demographics

without any preceding conditioning, is a practice that has been Characteristic N = 14


used with increasing frequency to treat PGF. Retrospective Diagnosis at HCT
studies in adult recipients of alloSCT have shown that hemato- ALL in CR1 1 (7%)
poietic recovery and survival are improved after CD34+SCB ALL in CR2 3 (21%)
[3,10-18], although the definition of PGF and measurements of
MDS/CMML 1 (7%)
success are not consistent throughout these studies.
MDS 1 (7%)
The available literature on outcomes after SCB is comprised
FA-MDS/AML 1 (7%)
almost exclusively from data on adult patients, whose predis-
FA-SAA 3 (21%)
posing diseases and transplant courses differ greatly from chil-
Anemia - SAA 2 (14%)
dren. To our knowledge, only 2 published studies of post-
CGD 1 (7%)
transplantation CD34+SCB for PGF include young pediatric
patients [19,20]. Both studies demonstrated an improvement Wiskott Aldrich syndrome 1 (7%)

of PGF in children and minimal toxicity including a low inci- Age at HCT 7.8 (0.9-24.5)
dence of acute or chronic GHVD. However, neither study Donor match code
included patients whose PGF was associated with a recent MRD 1 (7%)
infectious complication. Here we report on a single-center MUD 6 (43%)
experience with CD34+SCB for PGF after alloSCT on the Pediat- MMUD 7 (50%)
ric Transplant and Cellular Therapy Service at MSK Kids, Graft source at HCT
Memorial Sloan Kettering Cancer Center. Defining PGF more CD34+ PBSC 9 (64%)
broadly than published literature to include patients with con- CD34+ BM 1 (7%)
comitant infections, we demonstrate that CD34+SCB is safe Unmodified BM 4 (22%)
and can provide for durable trilineage hematopoietic recovery Regimen intensity at HCT
and long-term survival. MA 12 (86%)
RIC 2 (14%)
METHODS
Patients Primary HCT CD34+ cell dose/kg (106) 7.18 (3.46-16.57)
An Institutional Review Board approved retrospective analysis of data
Time to engraftment (ANC 500), d 12.0 (10.0-27.0)
was performed on all patients with malignant or nonmalignant diseases who
received a CD34+SCB on the Pediatric Stem Cell Transplantation and Cellular Time between primary HCT and boost (d) 148 (50-431)
Therapy Service at MSK Kids between 2008 to 2020 for PGF. PGF was defined Donor BM % chimerism 100 (86-100)
as the need for granulocyte colony stimulating factor (G-CSF) or packed red Donor T cell % chimerism 87 (0-100)
blood cells (PRBC) or platelet transfusion support with bone marrow donor
PGF associated with infection
chimerism 85% as assessed by short tandem repeat molecular testing in the
absence of recurrent disease for patients with leukemia. Thirteen patients Yes 8 (57%)
received G-CSF mobilized peripheral blood stem cells, and one patient CMV/adenovirus/HHV6 6
(patient 1) received bone marrow from the original donors with CD34 selec-
EBV 2
tion performed by the CliniMACS CD34 reagent system (Miltenyi Biotech,
Gladbach, Germany). There was no cap on the number of CD34+ cells/kg. Mycobacterium 1
However, the number of CD3+ T cells/kg was capped at 1 £ 104/kg. No patient No infection 6 (43%)
received additional immunosuppression or GVHD prophylaxis after CD34
Treatment before boost
+SCB infusion.
None 10 (71%)
Outcomes and Statistical Analyses ATG 3 (21%)
Time to PGF was determined by decline in ANC prompting use of G-CSF ATG, Fludarabine 1 (7%)
support, need for PRBC transfusion to maintain HB  7 g/dL, or in the case of
Boost CD34+ cell dose/kg (£ 106) 7.20 (3.14-19.07)
primary thrombocytopenia defined as failure to achieve a platelet count 
50,000 by day 60 or secondary thrombocytopenia defined as platelet count  ALL indicates acute lymphoblastic leukemia; AlloSCT, allogeneic stem cell
50,000 occurring after recovery (100,000) and persisting for more than transplantation; AML, acute myelogenous leukemia; ATG, antithymocyte glob-
30 days. For patients who had primary thrombocytopenia, the time to PGF ulin; CGD, chronic granulomatous disease; CMML, chronic myelomonocytic
was set at 60 days [5,21-23]. Complete recovery (CR) was defined as achieve- leukemia; CMV, cytomegalovirus; CR1, first complete remission; CR2, second
ment of an ANC  500 £ 106/L without need for G-CSF support and HB  complete remission; EBV, Epstein Barr virus; FA, Fanconi anemia; HCT,
7 g/dL and platelet count  50 £ 109/L without transfusion support by hematopoietic cell transplant; HHV6, human herpes virus 6; MA, myeloabla-
60 days after CD34+ SCB. Durable CR was defined as having achieved CR and tive; MDS, myelodysplastic syndrome; MRD, matched related donor; MUD,
remaining in CR at time of last follow-up. All patients were monitored for matched unrelated donor; MMUD, mismatched unrelated donor; PBSC,
development of GVHD. peripheral blood stem cells; RIC, reduced intensity conditioning; SAA, severe
aplastic anemia.
46.e3 E. Fraint et al. / Transplantation and Cellular Therapy 29 (2023) 46.e1 46.e6

associated with an infection. Patient diagnoses at initial SCT

Time to
SCB (d)
included malignant (n = 7) and nonmalignant (n = 7) disorders.

278
431
142
216
146
176
243
150

142

371
56

50

70
64
The median age of the patients was 7.8 years (range 0.9-24.5
years). SCT donors included related (n = 1) and unrelated
donors (n = 13); 7 of these SCT were HLA identical and 7 were
Time to
PGF (d)
HLA mismatched with matching defined at 10 alleles. For 12

unk
367
60

60
60
60
64
60

60
60
60
60
60
46
patients the alloSCT was their first alloSCT, and for 2 patients it
was their second alloSCT (patients no. 3 and no. 10).
Details of the SCT were not available for one patient
Plt >100k

(patient no. 8) whose SCT occurred at another institution. For


Time

the other thirteen patients, the median CD34+ cell dose for

unk
n/a

n/a
n/a
n/a

n/a

n/a
n/a
n/a
n/a
n/a
(d)

21

21

15
their alloSCT was 7.18 £ 106/kg (range 3.46-16.57 £ 106/kg).
All 13 patients had recovered an ANC  500 at a median of
12 days (range 10-27 days). Despite all patients experiencing
Plt >50k

neutrophil engraftment, only 5 patients recovered a platelet


Time

unk
n/a

n/a
n/a

n/a

n/a

n/a
n/a
n/a
(d)

count  50,000 by day 60 after initial SCT. Furthermore, only 3


21

48
21

43

15
patients recovered a platelet count  100,000 at any time prior
to the CD34+SCB. The median time to PGF was 60 days (range
Plt >20k

46-367 days [one outlier]). The median time between SCT and
Time

CD34+SCB was 148 days (range 50-431 days). Bone marrow


unk
216

n/a
n/a

n/a

n/a

n/a
(d)

20

13
21
30

37

99

15
biopsies were performed in 11 of 14 patients prior to CD34
+SCB and were notably hypocellular with a median of 10% cel-
lularity (range <5%-70%). The median donor bone marrow chi-
ANC 500

merism was 100% (range 86%-100%). The median donor T cell


Time

unk

chimerism was 87% (range 0%-100%).


(d)
15
13
10
17
11
12
11

22
10
12
10
27
11

At the time of PGF, 8 of 14 patients had at least one


associated infection: CMV/adenovirus/HHV6 (n = 6), EBV
CD34/kg

(n = 2), or mycobacterium (n = 1). Infections had resolved


(£106)

16.57

14.87
13.11

in 4 patients before the SCB. Two patients on foscarnet and


3.46
5.35
3.96
8.97
4.04

8.32

6.12
10.8
3.84
7.18
unk

1 patient on ethambutol at the time of SCB resolved their


M indicates male; F, female; d, days; P/D, patient/donor; n/a, not applicable; unk, unknown; WAS, Wiskott Aldridge syndrome.

infections after the SCB. One patient died of multiorgan


failure on day 10 with adenovirus infection. Three patients
(P/D)
CMV

unk
/+

/+
+/+
+/+

+/+
+/+
+/+

+/+

+/+

+/+

had single lineage PGF requiring either G-CSF, PRBC trans-


/

/
+/

fusions or platelet transfusions; 2 patients had bilineage


PGF, and 9 patients had trilineage PGF. Two patients with
MMUD

MMUD

MMUD

MMUD

MMUD

MMUD
MMUD

trilineage involvement had no response to romiplostim


Match
Donor

MUD
MUD
MUD

MUD

MUD

MUD
MRD

(patient 3) or eltrombopag (patient 4). Ten patients had no


lymphodepleting treatment prior to CD34+SCB; 8 of these
10 patients had 90% to 100% donor T-cell chimerism. Three
Unmodified BMT
Unmodified BMT
Unmodified BMT

Unmodified BMT

patients received ATG alone (donor T-cell chimerism 0%,


53%, and 66%) and 1 patient with 3% donor T-cell chime-
CD34+ PBSC
CD34+ PBSC
CD34+ PBSC

CD34+ PBSC
CD34+ PBSC
CD34+ PBSC
CD34+ PBSC
CD34+ PBSC
CD34+ PBSC
CD34+ BM

rism received ATG and fludarabine. The median SCB CD34+


Source of
Primary
alloSCT

cell dose was 7.20 £ 106/kg (range 3.14-19.07 £ 106). The


median SCB CD3+ cell dose cell was 3.44 £ 103/kg (range
0.45-8.82 x 103/kg). Nine patients received freshly collected
products and 5 patients received products that had been
Intensity

cryopreserved at the time of the primary alloSCT.


The probability of CR following CD34+SCB at 60 days after
MA

MA
MA
MA
MA
MA

MA
MA
MA
MA
MA
MA
RIC

RIC

CD34+SCB was 79% (95% CI, 57-100%; Figure 1A). The median
time to CR for 11 of 12 patients with CR was 20 days (range 7-
42 days). The twelfth patient with chronic granulomatous dis-
FA-MDS/AML

MDS/CMML

ease recovered hematopoietic function at day +166. All


Diagnosis

ALL CR2

ALL CR2

ALL CR1

ALL CR2
FA-SAA

FA-SAA

FA-SAA

patients with single or bilineage PGF and 7 of 9 patients with


WAS

MDS
CGD
SAA

SAA

trilineage PGF achieved CR (total 12 of 14 patients). Complete


recovery was durable as none of these patients required
subsequent growth factor or transfusion support in the
Individual Patient Demographics

Sex

absence of the development of autoimmune hemolytic ane-


M

M
M

M
M

M
F
F

mia (patient no. 10). Of the two patients with persistent


PGF, 1 had a successful second SCT (patient #2) and 1 died
13.6

14.5

24.5

10.1
20.6
20.5
Age
(yr)

of multiorgan failure (patient no. 3). Despite achieving CR,


7.2

0.9

3.3
3.8

6.8
7.8
7.8
7.3

1 patient died from the subsequent development of auto-


immune hemolytic anemia (patient no. 10) and one patient
died from acute GVHD (patient no. 13) that developed after
Patient
Table 2

CD34+SCB. Patient no. 13 was the only patient to develop


10
11
12
13
14
1
2
3
4
5
6
7
8
9

GVHD; 2 patients with a prior history of GVHD did not


Table 3
Individual Patient Evaluation and Outcome

Patient Infection Treatment for Infection h/o GVHD BM Cellularity BM Donor T cell Donor GCSF PRBC Tx Plt Tx Number Treatment SCB SCB Outcome
(%) chimerism (%) Chimerism (%) dep dep Lineages before SCB CD34/kg CD3/kg
Involved before SCB (£106) (£103)
1 CMV, Ganciclovir, Foscarnet, N 40 100 91 N Y N 1 none 4.32 0.45 CR Alive 64.9 mo
BK virus Cidofovir off treatment
2 EBV Rituximab Off treatment N 30 100 53 Y Y Y 3 ATG 16.08 3.26 PGF 2nd SCT Alive 99.4 mo
3 Adenovirus Cidofovir On treatment Y <5 95 100 Y Y Y 3 none 5.65 6.86 PGF died day 10 MOF
4 CMV, Ganciclovir, Foscarnet, Y 60 96 98 Y Y Y 3 none 9.85 3.00 CR alive 65.6 mo
Adenovirus Cidofovir Off treatment
5 CMV, Foscarnet On N <5 100 100 Y N Y 2 none 3.14 2.96 CR alive 119.8 mo
Adenovirus, maintenance
RSV

E. Fraint et al. / Transplantation and Cellular Therapy 29 (2023) 46.e1 46.e6


6 EBV, CMV Rituximab, Ganciclovir, N nd nd 12 Y N N 1 none 18 3.62 CR alive 55.4 mo
Foscarnet, EBV-Cytotoxic
T cells Off treatment
7 Mycobacterium Ethambutol N 70 100 100 Y Y Y 3 none 4.16 7.23 CR alive 119.8 mo
On treatment
8 CMV, HHV6 Ganciclovir, Foscarnet N nd nd nd Y Y Y 3 none 5.27 5.93 CR alive 18.9 mo
CMV-Cytotoxic
T cells On treatment
9 None None N 40 100 87 N Y Y 2 none 3.7 3.01 CR Alive 29.7 mo
10 None None N <5% 100 24 Y Y Y 3 none 19.07 3.92 CR died day 702 AIHA
11 None None N 9 88 0 Y Y Y 3 ATG 4.05 4.25 CR alive 34.6 mo
12 None None N nd 100 95 N N Y 1 none 10.04 8.82 CR alive 109.5 mo
13 None None N 10 88 66 Y Y Y 3 ATG 8.97 2.72 CR died day 315 GVHD
14 None None N 9 86 3 Y Y Y 3 ATG, 8.74 2.64 CR alive 60.3 mo
fludarabine

AIHA indicates autoimmune hemolytic anemia; h/o, history of; nd, not done; MOF multi-organ failure; PRBC Tx dep, packed red blood cell transfusion dependent; Plt Tx dep, platelet transfusion dependent; RSV, respiratory syncytial
virus.

46.e4
46.e5 E. Fraint et al. / Transplantation and Cellular Therapy 29 (2023) 46.e1 46.e6

survival

Figure 1. Results in patients receiving CD34-selected stem cell boost. (A) The cumulative incidence of complete recovery by 60 days after CD34+SCB was 79% (95% CI,
57%-100%). (B) The overall survival after CD34+SCB at both 2 years and 5 years was 78% (95% CI, 56%-100%).

experience a flare of GVHD after CD34+SCB. No additional involvement were included in our study, because the long-
toxicity related to CD34+SCB was noted. term need for G-CSF support, PRBC transfusions, or platelet
Six patients had recovered an absolute CD4+ T cell count transfusions can lead to increased morbidity and NRM. Our
>200 cells/mL before the SCB. Eight patients had not immune study demonstrates the wide applicability of therapeutic CD34
reconstituted before the SCB; 5 patients who achieved CR and +SCB, as all those with single (n = 3) or bilineage (n = 2) PGF,
survived experienced full immune reconstitution whereas the and 7 of 9 patients with trilineage PGF had CR (total 12 of 14
3 patients who died did not immune reconstitute before patients).
death. Some prior literature with adult patients demonstrates
The overall survival following CD34+SCB at both 2 and similar outcomes to our cohort. Shahzad et al. [24] published a
5 years was 78% (95% CI, 56%-100%; Figure 1B). The median fol- systematic review and meta-analysis of 7 studies with 209
low-up time from SCB was 60.3 months (range 3.4 months to adult patients with leukemia who received CD34+SCB for
10.0 years). treatment of PGF. In their meta-analysis, the median time
from alloSCT to CD34+SCB was 138 days (range 113-450 days),
DISCUSSION like the time period reported in our series of 163 days (range
In this retrospective study, we report excellent survival 50-431 days). Similarly, their complete hematologic response
with very low toxicity in pediatric recipients of CD34+SCBs for rate was 72% (95% CI, 63%-79%), which is analogous to the CR
treatment of PGF, about half of whom had an associated infec- rate of 79% (95% CI, 57%-100%) in our study. For the 209 adult
tion. Hematologic responses were common with a CR rate of patients, OS ranged from 80% at 1 year to 40% at 9 years with
79% and occurred at a median of 20 days after CD34+SCB, lead- NRM of 27% and death due to relapse in 17% of patients. In our
ing to excellent OS of 78% at 5 years. These 14 patients demon- series, the OS was 78% (95% CI, 56%-100%) at 5 years. Because
strated a markedly improved outcome compared to the of the heterogeneity of the adult patients in the study by Shah-
historically abysmal fate of patients with PGF [7 9]. Overall, zad et al. [24], pooled analysis based on donor type and other
CD34+SCBs were well tolerated; two patients with a prior his- parameters could not be done. However, the authors con-
tory of aGVHD did not have a flare of aGVHD (patients no. 3 cluded that CD34+SCB infusion could provide a safe and effec-
and no. 4), and only 1 patient developed any level of GVHD, tive treatment option in adult patients with PGF after alloSCT.
although it was fatal. Literature describing pediatric patients is more limited.
Existing literature has defined PGF variably, using different Most notably, Mainardi et al. [19] reported a single-center
thresholds for cytopenias and requiring different numbers of study of CD34+SCBs that included pediatric patients (n = 50,
affected lineages, with most authors describing at least 2 median age 11.8 years, range 0.33-38.98 years). They included
affected lineages for inclusion [6]. However, most recently, patients with at least bilineage cytopenias or dependence on
ASTCT definitions have been published that do not specify transfusion with full donor chimerism in the absence of dis-
multilineage involvement [4]. Although most of our patients ease relapse or GVHD. Concomitant infection in these patients
had 2- or 3-lineage involvement, 3 patients with single-lineage was not addressed. Although 78.8% of their patients had an
E. Fraint et al. / Transplantation and Cellular Therapy 29 (2023) 46.e1 46.e6 46.e6

improvement in 1 or 2 lineages, only 36% achieved complete REFERENCES


hematologic recovery. Their 5-year OS was low (38.67%) 1. Kong Y. Poor graft function after allogeneic hematopoietic stem cell trans-
plantation-an old complication with new insights. Semin Hematol.
because of NRM (24% of total) and relapse (28% of total). It is 2019;56:215–220.
not clear why they had such different results from our study, 2. Zhao Y, Gao F, Shi J, et al. Incidence, risk factors, and outcomes of primary
but there are some notable differences between our patient poor graft function after allogeneic hematopoietic stem cell transplanta-
tion. Biol Blood Marrow Transplant. 2019;25:1898–1907.
populations. The outsized contribution of patients with malig-
3. Chen J, Wang H, Zhou J, Feng S. Advances in the understanding of poor
nant disorders in their cohort (nearly all their patients, with graft function following allogeneic hematopoietic stem-cell transplanta-
almost 30% not in remission at the time of initial transplanta- tion. Ther Adv Hematol. 2020;11: 2040620720948743.
4. Kharfan-Dabaja MA, Kumar A, Ayala E, et al. Standardizing definitions of
tion) as compared with our cohort, only half of whom had
hematopoietic recovery, graft rejection, graft failure, poor graft function,
malignancy, likely plays a role in these divergent outcomes, and donor chimerism in allogeneic hematopoietic cell transplantation: a
because relapse led to a large proportion of their patient report on behalf of the American Society for Transplantation and Cellular
deaths. Our study demonstrates that CD34+SCB can be both Therapy. Transplant Cell Ther. 2021;27:642–649.
5. Prabahran A, Koldej R, Chee L, Wong E, Ritchie D. Evaluation of risk factors
effective and well tolerated in pediatric patients with malig- for and subsequent mortality from poor graft function (PGF) post alloge-
nant or nonmalignant disorders experiencing PGF. neic stem cell transplantation. Leuk Lymphoma. 2021;62:1482–1489.
The incidence of acute GVHD among the 209 adult 6. Prabahran A, Koldej R, Chee L, Ritchie D. Clinical features, pathophysiol-
ogy, and therapy of poor graft function post-allogeneic stem cell trans-
recipients of CD34+SCB in the meta-analysis by Shahzad plantation. Blood Adv. 2022;6:1947–191959.
et al. [24] was 17% (95% CI, 13%-23%) [24].In our small 7. Guardiola P, Pasquini R, Dokal I, et al. Outcome of 69 allogeneic stem cell
pediatric series, only one patient developed any level of transplantations for Fanconi anemia using HLA-matched unrelated
donors: a study on behalf of the European Group for Blood and Marrow
aGVHD, although it was fatal. No additional toxicities Transplantation. Blood.. 2000;95:422–429.
related to the CD34+SCB were observed in any other 8. Chewning JH, Castro-Malaspina H, Jakubowski A, et al. Fludarabine-based
patients. Using CD34+SCB as a tool to correct PGF after conditioning secures engraftment of second hematopoietic stem cell
allografts (HSCT) in the treatment of initial graft failure. Biol Blood Marrow
alloSCT is attractive, especially in comparison to the option
Transplant. 2007;13:1313–1323.
of repeat SCT. Repeat alloSCT after preparative conditioning 9. Jabbour E, Rondon G, Anderlini P, et al. Treatment of donor graft failure
can successfully improve PGF with response rates of 66% to with nonmyeloablative conditioning of fludarabine, antithymocyte globu-
lin and a second allogeneic hematopoietic transplantation. Bone Marrow
100%, but this comes at the expense of additional exposure
Transplant. 2007;40:431–435.
to cytotoxic medications with inherent short and long-term 10. Oyekunle A, Koehl U, Schieder H, et al. CD34(+)-selected stem cell boost
associated risks and toxicities, GVHD, infectious complica- for delayed or insufficient engraftment after allogeneic stem cell trans-
tions, and other toxicities that contribute to NRM. plantation. Cytotherapy. 2006;8:375–380.
11. Larocca A, Piaggio G, Podesta M, et al. Boost of CD34+-selected peripheral
Our study is limited by the single institution focus, small blood cells without further conditioning in patients with poor graft func-
sample size and retrospective nature, but it provides insight tion following allogeneic stem cell transplantation. Haematologica.
on the utility of CD34+SCB for ameliorating cytopenias in pedi- 2006;91:935–940.
12. Klyuchnikov E, El-Cheikh J, Sputtek A, et al. CD34(+)-selected stem cell
atric patients with PGF post alloSCT. Our cohort demonstrates boost without further conditioning for poor graft function after allogeneic
the use of CD34+SCB in a wide range of patient types, including stem cell transplantation in patients with hematological malignancies.
those transplanted for malignant and nonmalignant disorders; Biol Blood Marrow Transplant. 2014;20:382–386.
13. Askaa B, Fischer-Nielsen A, Vindelov L, Haastrup EK, Sengelov H. Treat-
with myeloablative or reduced intensity conditioning; with ment of poor graft function after allogeneic hematopoietic cell transplan-
either HLA matched or HLA mismatched donors; and with con- tation with a booster of CD34-selected cells infused without conditioning.
comitant viral or mycobacterial infections. Our cohort is too Bone Marrow Transplant. 2014;49:720–721.
14. Stasia A, Ghiso A, Galaverna F, et al. CD34 selected cells for the treatment
small to answer questions about whether immunosuppression
of poor graft function after allogeneic stem cell transplantation. Biol Blood
prior to CD34+SCB improves outcomes, but most of the Marrow Transplant. 2014;20:1440–1443.
patients here, as in other studies, did not receive cytoreduction 15. Haen SP, Schumm M, Faul C, Kanz L, Bethge WA, Vogel W. Poor graft func-
tion can be durably and safely improved by CD34+-selected stem cell
or lymphodepletion before the infusion of CD34+SCB.
boosts after allogeneic unrelated matched or mismatched hematopoietic
Although PGF is not a common complication of transplanta- cell transplantation. J Cancer Res Clin Oncol. 2015;141:2241–2251.
tion, exploring best practices in treatment of PGF is important 16. Ghobadi A, Fiala MA, Ramsingh G, et al. Fresh or cryopreserved CD34
because of the high morbidity and mortality associated with (+)-selected mobilized peripheral blood stem and progenitor cells for the
treatment of poor graft function after allogeneic hematopoietic cell trans-
PGF. This study demonstrates that for pediatric allogeneic SCT plantation. Biol Blood Marrow Transplant. 2017;23:1072–1077.
recipients with PGF and high donor bone marrow chimerism 17. Mohty R, Brissot E, Battipaglia G, et al. CD34(+)-selected stem cell "Boost"
of at least 85%, whether idiopathic or associated with an infec- for poor graft function after allogeneic hematopoietic stem cell transplan-
tation. Curr Res Transl Med. 2019;67:112–114.
tion, CD34-selected peripheral blood stem cell boost is safe 18. Cuadrado MM, Szydlo RM, Watts M, et al. Predictors of recovery following
and can provide for durable trilineage engraftment and long- allogeneic CD34+-selected cell infusion without conditioning to correct
term survival. poor graft function. Haematologica. 2020;105:2639–2646.
19. Mainardi C, Ebinger M, Enkel S, et al. CD34(+) selected stem cell boosts can
improve poor graft function after paediatric allogeneic stem cell trans-
plantation. Br J Haematol. 2018;180:90–99.
ACKNOWLEDGMENTS 20. Berger M, Faraci M, Saglio F, et al. CD34+ selected peripheral blood Stem
Cell Boost (SCB) for Poor Graft Function (PGF) or mixed chimerism in pedi-
The authors acknowledge the editorial assistance of Joe
atric patients, after hematopoietic stem cell transplantation: Results of a
Olechnowicz, Editor, MSK Kids. retrospective multicenter study. Pediatr Transplant. 2021;25(5):e13909.
Financial disclosure: Supported by grants from the NIH/NCI 21. Kim DH, Sohn SK, Baek JH, et al. Clinical significance of platelet count at
day +60 after allogeneic peripheral blood stem cell transplantation. J
Cancer Center Support Grant P30 CA008748.
Korean Med Sci. 2006;21:46–51.
Conflict of interest statement: There are no conflicts of inter- 22. Tang FF, Sun YQ, Mo XD, et al. Incidence, risk factors, and outcomes of primary
est to report. prolonged isolated thrombocytopenia after haploidentical hematopoietic
stem cell transplant. Biol Blood Marrow Transplant. 2020;26:1452–1458.
Authorship statement: All authors provided contributions to
23. Zaja F, Geromin A, Patriarca F, et al. Prognostic significance of delayed
the concept or design of the work; or the acquisition, analysis, thrombocytopenia after allogeneic stem cell transplant. Am J Hematol.
or interpretation of data for the work. E.F. and N.A.K. wrote the 2011;86:790–792.
24. Shahzad M, Siddiqui RS, Anwar I, et al. Outcomes with CD34-selected stem
initial drafts and all authors provided critical review and revi-
cell boost for poor graft function after allogeneic hematopoietic stem cell
sions for intellectual content. All authors approved the submit- transplantation: a systematic review and meta-analysis. Transplant Cell
ted version. Ther. 2021;27(10):877.. e1- e8.

You might also like