1 s2.0 S0006497121069950 Main
1 s2.0 S0006497121069950 Main
1 s2.0 S0006497121069950 Main
TRANSPLANTATION
KEY POINTS Patients with B-lineage acute lymphoblastic leukemia (ALL) are at high-risk for relapse after
allogeneic hematopoietic cell transplantation (HCT). We conducted a single-center phase 2
Blinatumomab is safe
and feasible for use in study evaluating the feasibility of 4 cycles of blinatumomab administered every 3 months
B-ALL after allogeneic during the first year after HCT in an effort to mitigate relapse in high-risk ALL patients.
HCT.
Twenty-one of 23 enrolled patients received at least 1 cycle of blinatumomab and were
The composition of a included in the analysis. The median time from HCT to the first cycle of blinatumomab was
patient’s T-cell subsets 78 days (range, 44 to 105). Twelve patients (57%) completed all 4 treatment cycles. Neutro-
at the time of treat- penia was the only grade 4 adverse event (19%). Rates of cytokine release (5% G1) and neu-
ment is indicative of
whether they will rotoxicity (5% G2) were minimal. The cumulative incidence of acute graft-versus-host disease
respond to (GVHD) grades 2 to 4 and 3 to 4 were 33% and 5%, respectively; 2 cases of mild (10%) and
blinatumomab. 1 case of moderate (5%) chronic GVHD were noted. With a median follow-up of 14.3
months, the 1-year overall survival (OS), progression-free survival (PFS), and nonrelapse mor-
tality (NRM) rates were 85%, 71%, and 0%, respectively. In a matched analysis with a contemporary cohort of 57
patients, we found no significant difference between groups regarding blinatumomab’s efficacy. Correlative studies of
baseline and posttreatment samples identified patients with specific T-cell profiles as “responders” or “nonresponders”
to therapy. Responders had higher proportions of effector memory CD8 T-cell subsets. Nonresponders were T-cell defi-
cient and expressed more inhibitory checkpoint molecules, including T-cell immunoglobulin and mucin domain 3 (TIM3).
We found that blinatumomab postallogeneic HCT is feasible, and its benefit is dependent on the immune milieu at time
of treatment. This paper is posted on ClinicalTrials.gov, study ID: NCT02807883.
BLINATUMOMAB IN B-ALL POST–ALLOGENEIC HCT blood® 24 MARCH 2022 | VOLUME 139, NUMBER 12 1909
Table 1. Patient and treatment characteristics (BD Biosciences: Clone MH1), CD19 (BD Biosciences: Clone
HIB19), TIM-3 (BD Biosciences: Clone 7D3), and TIGIT (eBio-
Measure All patients, n 5 21, (%) science: Clone MBSA43). Cells were acquired using an X-20 For-
tessa (BD Biosciences). The analysis was implemented with the
Gender
cyt3 package in MATLAB (https://github.com/dpeerlab/cyt3).24
Male 17 (81) Raw data were first transformed using hyperbolic arcsin with a
Female 4 (19) cofactor of 150. Bh-SNE25 version of SNE was run against the
collection of all samples, with 8000 data points subsampled for
Age
each; viSNE reduced the high-dimensional flow cytometry data
Mean (SD) 37.9 (16.2) into 2 dimensions, and the result was visualized in the viSNE
Median (range) 29.9 (16.9-65.6) map (2D scatterplot). PhenoGraph clustering26 was then used to
identify subpopulations in the viSNE map. The number of near-
Race/ethnicity
est neighbors was set at 100. The annotation of subpopulations
White 13 (62) by PhenoGraph was directly shown in the viSNE map, where
Hispanic 8 (38) cells that belong to the same subpopulation were shown in the
same color.
High-risk cytogenetic/ 18 (90)
molecular risk at diagnosis
Response definitions and outcome
Ph1 2
CR was defined as having #5% malignant blasts in the BM, nor-
Ph-like 8
mal blood counts with ANC $0.5 3 109/L and platelet .20 3
KMT2A 4 109/L, normal karyotype, and absence of extramedullary disease.
Complex* 2 MRD was assessed using multiparameter flow cytometry with a
Hypodiploid 2 threshold of .0.01%. Myeloablative and reduced-intensity con-
ditioning regimens were defined according to the Center for
Days from HCT to International Blood and Marrow Transplant Research (CIBMTR)
blinatumomab start
criteria.27 aGVHD was staged and graded according to the crite-
Median (range) 78.0 (44.0-105.0) ria published by Przepiorka and colleagues.28 Chronic GVHD
MRD prior to blinatumomab
(cGVHD) was reported based on the 2014 NIH cGVHD Consen-
treatment sus Conference.29
Detected 2 (10)
Statistical methods
Not detected 19 (90)
OS time and NRM were computed from date of allogeneic HCT
Number of cycles to last known vital sign. Patients alive at the last follow-up date
1 21 were censored. PFS time was computed from date of allogeneic
HCT to date of disease progression or death (if he or she died
2 13
without disease progression) or the last evaluation date. Patients
3 12
who were alive and did not experience progression of disease
4 12 at the last follow-up date were censored. In addition, relapse
and GVHD were computed from date of allogeneic HCT to date
*Complex karyotype defined as 5 or more cytogenetic abnormalities. of event; patients who did not experience the event were cen-
sored at their last follow-up date. Patients who remained in
and at study completion. Patients who remained in remission remission were grouped as “responders,” and those that pro-
after blinatumomab therapy were labeled “Responders,” and gressed during therapy with blinatumomab were grouped as
patients with disease progression defined by recurrence of MRD “nonresponders.” OS and PFS were estimated using the
by flow cytometry or morphologic leukemic blasts were labeled Kaplan-Meier method. The cumulative incidences of relapse,
“Nonresponders.” The Common Terminology Criteria for NRM, and GVHD were evaluated using the competing risks
Adverse Events (CTCAE) version 4 was used to grade toxicities. method. The competing risk for relapse was death and for NRM
was relapse, while the competing risks for GVHD were relapse
Flow cytometry data analysis and death.
Samples were collected following informed consent from 15
consecutive patients (4 nonresponders, 11 responders), sepa- A contemporary cohort of patients with high-risk B-ALL who
rated using Ficoll density separation (Lymphoprep, STEMCELL received allogeneic HCT was identified retrospectively. To cor-
Technologies), and cryopreserved. PB mononuclear cells (PBMC) rect for potential bias in the HCT comparisons, treatment
were then thawed and immunostained with CD127 (BD Bio- patients were matched to controls (1:2 where possible) using
sciences: Clone HIL7RM21), CD25 (BioLegend: Clone BC96), the following steps: 1) patients were divided into groups based
LAG-3 (BD Biosciences: Clone T47-530), PD-1 (BioLegend: on disease status at allogeneic HCT, cytogenetic risk, and MRD
Clone EH12.2H7), CCR7 (BD Biosciences: Clone 3D12), CD3 prior to allogeneic HCT; we did not match for conditioning regi-
(BD Biosciences: Clone UCHT1), CD45RO (BD BioSciences: men intensity as a separate analysis comparing the 2 treatment
Clone UCHL1), CD8 (BioLegend: Clone SK1), 2B4 (BioLegend: groups by regimen intensity did not yield different results (data
Clone C1.7), CTLA-4 (BioLegend: Clone L3D10), CD160 (BioLe- not shown); 2) within each subgroup of completely matched
gend: Clone 341204), CD4 (BioLegend: Clone RPA-T4), PDL1 patients, a random number generator was employed from the
HCT patients
Blinatumomab
Measure All, n 5 57 (%) n 5 21 (%) Controls n 5 36 (%) P value*
Gender
Male 33 (58) 17 (81) 16 (44) .012
Female 24 (42) 4 (19) 20 (56)
Age in years, median 38.0 (16.0-66.0) 29.0 (16.0-65.0) 41.0 (19.0-66.0) .26†
(range)
Race/ethnicity
White 36 (65) 13 (62) 23 (68) .13
Hispanic 15 (27) 8 (38) 7 (21)
Other 4 (7) 0 (0) 4 (12)
MRD at HCT
Detected 10 (18) 4 (20) 6 (17) .73
Not detected 46 (82) 16 (80) 30 (83)
Unknown 1 1 0
Karnofsky performance
status
#80 5 (11) 1 (6) 4 (13) .65
$80 42 (89) 15 (94) 27 (87)
HCT-CI
Median (Range) 3.0 (0.0-8.0) 3.0 (0.0-8.0) 3.0 (0.0-8.0) .91†
Donor type
MRD 22 (39) 7 (33) 15 (42) .48
MUD 21 (37) 10 (48) 11 (31)
Haplo 14 (25) 4 (19) 10 (28)
Conditioning regimen
MAC 25 (44) 4 (19) 21 (58) .010
MAC/TBI 8 (14) 5 (24) 3 (8)
RIC 24 (42) 12 (57) 12 (33)
uniform distribution on the interval (0,1) using a prime modulus while group differences in cumulative incidences were assessed
multiplicative generator; and 3) each treated patient was by stratified Gray’s test.30
matched with 1 or 2 (where possible) control patient(s) starting
from the lowest generated random number. Group differences All statistical analyses were performed using SAS 9.4 for Win-
in OS and PFS were evaluated using a stratified log-rank test dows (SAS Institute Inc., Cary, NC). All statistical tests used a
BLINATUMOMAB IN B-ALL POST–ALLOGENEIC HCT blood® 24 MARCH 2022 | VOLUME 139, NUMBER 12 1911
Table 3. Incidence of toxicities graded by CTCAE, V4
Maximum grade
Hematologic
Anemia 1 (5) 0 (0) 0 (0) 0 (0)
Leukopenia 2 (10) 6 (29) 4 (19) 0 (0)
Neutropenia 2 (10) 0 (0) 0 (0) 4 (19)
Thrombocytopenia 3 (14) 0 (0) 1 (5) 0 (0)
Cardiovascular
Chest pain 0 (0) 1 (5) 0 (0) 0 (0)
Hypotension 0 (0) 1 (5) 0 (0) 0 (0)
Arrythmias 1 (5) 0 (0) 0 (0) 0 (0)
Thromboembolic event 2 (10) 0 (0) 0 (0) 0 (0)
Pulmonary
Dyspnea 0 (0) 1 (5) 0 (0) 0 (0)
Cough 1 (5) 0 (0) 0 (0) 0 (0)
Gastrointestinal
Infections
Viral 2 (10) 0 (0) 0 (0) 0 (0)
Bacterial 1 (5) 0 (0) 0 (0) 0 (0)
Electrolyte abnormalities
Hypokalemia 0 (0) 0 (0) 1 (5) 0 (0)
Hypomagnesemia 1 (5) 0 (0) 0 (0) 0 (0)
Neurologic
Headache 3 (14) 0 (0) 0 (0) 0 (0)
Dizziness 1 (5) 0 (0) 0 (0) 0 (0)
Confusion 0 (0) 1 (5) 0 (0) 0 (0)
Transient dysphasia 1 (5) 0 (0) 0 (0) 0 (0)
Tremors 1 (5) 0 (0) 0 (0) 0 (0)
Skin
Rash 4 (19) 1 (5) 2 (10) 0 (0)
Renal
Elevated creatinine 1 (5) 1 (5) 0 (0) 0 (0)
Maximum grade
significance level of 5%. No adjustments for multiple testing patients were in CR1, and 80% had no detectable MRD at the
were made. time of HCT. The Karnosky performance score was $80% in
94% of patients, and the median comorbidity index was 3
(range, 0 to 8). Regarding the source of allogeneic HCT graft,
Results 33% were matched siblings, 48% matched unrelated donors,
Patient and disease characteristics and 19% had haploidentical family donors. Approximately half
Twenty-three patients signed consent, and 21 patients who of the patients received a reduced-intensity conditioning regi-
received at least 1 dose of blinatumomab therapy postalloge- men. The median time from diagnosis to HCT was 8.8 months
neic HCT were included in the analysis. Two patients never (range, 3.2 to 107.7 months). The median number of blinatumo-
received therapy due to GVHD that required treatment. Table mab cycles received was 4 (range, 1 to 4). Except for gender, all
1 summarizes patient and treatment characteristics for the key characteristics were similar between the study and control
study cohort. Eighty-one percent of the patients were male, groups (Table 2).
62% were White, with a median age of 30 (17 to 66) years, and
90% (19 of 21) had a high-risk cytogenetic or molecular profile Safety and feasibility
at diagnosis. Two patients had .1 HCT prior to blinatumomab Blinatumomab was well-tolerated posttransplantation, with the
therapy. Seventy-six percent of patients were exposed to blina- most common severe adverse events being limited to hemato-
tumomab prior to allogeneic HCT. The median days from trans- logic cytopenias, including leukopenia (19% G3) and neutrope-
plant to the first day of cycle 1 of blinatumomab was 78 (range, nia (19% G4), as noted in Table 3. Diarrhea occurred in 7
44 to 105), and MRD was detected prior to the start of blinatu- patients (33%) and was mostly grade 1 (5 patients) and not
momab in 2 patients. Fifty-seven percent of patients (12 of 21) GVHD-related. Importantly, only 1 patient developed grade 1
completed all 4 cycles of therapy (Table 1). Three patients CRS, and 1 patient developed grade 2 neurotoxicity in the form
could not complete treatment due to GVHD, and the remain- of confusion that resolved with a temporary hold of the blinatu-
ing patients (n 5 6) relapsed before they could complete all 4 momab infusion (Table 3). Furthermore, rates of GVHD were
intended cycles. All patients were on tacrolimus during cycle 1 acceptable, with cumulative rates of aGVHD grades 2 to 4 and
of blinatumomab, with a mean tacrolimus level of 7.4 ng/mL 3 to 4 noted at 33% and 5%, respectively. Two patients (10%)
(range, 4.3 to 10.3 ng/mL). were noted to have NIH mild cGVHD (oral 1/3; oral 1/3, liver
1/3), and 1 patient (5%) had moderate cGVHD (skin 3/3, liver
Transplant characteristics for the study group and the matched 1/3). Finally, none of the patients developed secondary graft fail-
cohort are listed in Table 2. In the study group, about half of the ure. Study accrual was slow, and consequently, the study was
Baseline High
Mean arcsinh-transformed
Non-responder 1.5
fluorescent intensity
Responder
1
Posttreatment
Non-responder 0.5
Responder
0
Low
8
3
1
R7
CD D3
CD O
0
L1
4
IT
3
19
4
CD
2B
G
PD
16
CD
LA
R
G
PD
CD
CC
C
LA
45
TI
TI
CT
Figure 1. Heatmap of surface marker expression of T cells in nonresponders and responders. Hyperbolic arcsin transformed fluorochrome expression for 14
markers were averaged for baseline samples taken from nonresponders (n 5 3) and responders (n 5 10), and posttreatment samples from nonresponders (n 5 4) and
responders (n 5 11).
BLINATUMOMAB IN B-ALL POST–ALLOGENEIC HCT blood® 24 MARCH 2022 | VOLUME 139, NUMBER 12 1913
A Non-responders Responders
15 15
10 10
5 5
bh-SNE2
bh-SNE2
Baseline
0 0
–5 –5
–10 –10
–15 –15
–15 –10 –5 0 5 10 15 –15 –10 –5 0 5 10 15
bh–SNE1 bh–SNE1
15 15
10 10
Posttreatment
5 5
bh-SNE2
bh-SNE2
0 0
–5 –5
–10 –10
–15 –15
–15 –10 –5 0 5 10 15 –15 –10 –5 0 5 10 15
bh–SNE1 bh–SNE1
CD8 T cells: 2 17 19 22
CD4 T cells: 5 12 16 25
1 4 7 10 13 16 19 22 25
2 5 8 11 14 17 20 23
3 6 9 12 15 18 21 24
B
Cluster
1 (10.32%)
High
2 (9.99%)
5
Mean arcsinh-transformed
3 (9.83%)
fluorescent intensity
4 (9.59%) 4
5 (7.80%)
3
6 (6.40%)
7 (6.25%) 2
8 (5.42%)
1
9 (5.04%)
10 (3.70%) 0
11 (3.67%) Low
12 (3.39%)
13 (3.20%)
14 (2.93%)
15 (2.93%)
16 (2.09%)
17 (1.69%)
18 (1.65%)
19 (1.23%)
20 (1.17%)
21 (0.93%)
22 (0.27%)
23 (0.26%)
24 (0.13%)
25 (0.13%)
8
LA B4
3
CC 1
CD CD7
45 3
CD RO
PD60
CD1
TI 4
TI IT
CD 3
CT 19
4
CD
G
PD
R
LA
G
1
2
Figure 2. Subpopulations identified via viSNE analysis of 14 surface markers in all 56 samples. (A) viSNE map for nonresponders and responders color-coded
according to PhenoGraph cluster annotation. viSNE maps were separated to baseline and posttreatment in both nonresponders and responders groups. (B) Heatmap
of mean surface marker expression in each cluster. Percentage in parentheses denotes the size of each cluster.
0.8
Laboratory correlates
Cumulative incidence of relapse
0.8 globulin and mucin domain 3), and CTLA4. Checkpoint mole-
cules LAG3, PD1, TIGIT, and TIM3 were expressed at high
++ ++
+
+ ++ + + ++ + +
levels both at baseline and after treatment in all patient samples
0.6 (Figure 1); however, TIM3 was the only checkpoint that was
expressed at statistically higher levels in nonresponders com-
pared with responders after blinatumomab treatment (P 5 .04)
0.4 (supplemental Figure 1). Finally, CD19 expression was lowest
after 1 cycle of blinatumomab in responders compared with
nonresponders (Figure 1), and the difference was statistically sig-
0.2 nificant after treatment (supplemental Figure 1).
Efficacy
Blinatumomab
Seventeen of the 21 (81%) patients were alive at the end of the
0.0
study, and the median follow-up time for all patients was 14.3
0 10 20 30 40 50 months (range, 7.5 to 52.4 months). Six patients progressed,
Months post HCT including the 2 patients who had MRD positivity prior to the
21 13 5 4 1 1 start of blinatumomab therapy, for a cumulative incidence of
C relapse of 29% (95% CI 11%-49%). The 1-year OS and PFS for
1.0
patients were 85% (95% CI 61%-95%) and 71% (95% CI 47%-
++ 86%), respectively (Figure 3). There were no regimen-related
0.8 +++
+ ++ + + ++ ++ + deaths. We compared our results to a contemporary cohort con-
trol that included information for 128 patients (Table 2). Using a
Probability of overall survival
0.2 Discussion
To our knowledge, this is the first study to investigate the use
Blinatumomab of prophylactic blinatumomab in the posttransplant setting to
0.0
0 10 20 30 40 50
Figure 3. Study outcomes for patients treated with blinatumomab. At 1 year,
Months post HCT the rate of relapse was 29% (95% CI, 11%-49%) (A), progression-free survival
21 16 6 5 2 1 (PFS) 71% (95% CI, 47%-86%) (B), and overall survival (OS) 85% (95% CI, 61%-
95%) (C).
BLINATUMOMAB IN B-ALL POST–ALLOGENEIC HCT blood® 24 MARCH 2022 | VOLUME 139, NUMBER 12 1915
Notably, we were able to glean important mechanistic insights
A into why this type of cellular therapy had benefits in only a sub-
1.0
set of patients. Broadly, cluster analysis clearly identifies res-
+ ponders as having higher frequencies of CD4 and CD8 T cells
Probability of progression-free survival
++
+ ++ ++ after cycle 3, specifically with an increase in the CD3/CD8 T-cell
++
subset. Furthermore, they noted increases in CD4/CD45RO1
0.6 + +
+ ++ + ++ ++
T-NK and NK lymphocyte populations, while they noted a pro-
gressive reduction in T-regulatory (Tregs) CD41 T cells, which
0.4 have been shown in some studies to drive tumor evasion and
limit the efficacy of blinatumomab.33,35,36 In contrast to our
study, they did not find a correlation between immune modula-
0.2
tion and the degree of molecular response that was reached in
Blinatumomab about 80% of patients.34
p=0.23
0.0 Controls
REFERENCES final results of the International ALL Trial pre-imatinib era: results from the Interna-
1. Goldstone AH, Richards SM, Lazarus HM, (MRC UKALL XII/ECOG E2993). Blood. tional ALL Trial MRC UKALLXII/ECOG2993.
2008;111(4):1827-1833. Blood. 2009;113(19):4489-4496.
et al. In adults with standard-risk acute lym-
phoblastic leukemia, the greatest benefit is 2. Fielding AK, Rowe JM, Richards SM, et al. 3. G€okbuget N, Kneba M, Raff T, et al;
achieved from a matched sibling allogeneic Prospective outcome data on 267 German Multicenter Study Group for
transplantation in first complete remission, unselected adult patients with Philadelphia Adult Acute Lymphoblastic Leukemia.
and an autologous transplantation is less chromosome-positive acute lymphoblastic Adult patients with acute lymphoblastic
effective than conventional consolidation/ leukemia confirms superiority of allogeneic leukemia and molecular failure display
maintenance chemotherapy in all patients: transplantation over chemotherapy in the a poor prognosis and are candidates for
BLINATUMOMAB IN B-ALL POST–ALLOGENEIC HCT blood® 24 MARCH 2022 | VOLUME 139, NUMBER 12 1917
stem cell transplantation and targeted hematologic neoplasms. N Engl J Med. 315-pre-hct-preparative-regimen-
therapies. Blood. 2012;120(9): 1989;320(13):828-834. conditioning.
1868-1876.
15. Miyamoto T, Fukuda T, Nakashima M, et al. 28. Przepiorka D, Weisdorf D, Martin P, et al.
4. Zhou Y, Slack R, Jorgensen JL, et al. The Donor lymphocyte infusion for relapsed 1994 Consensus conference on acute GVHD
effect of peritransplant minimal residual hematological malignancies after unrelated grading. Bone Marrow Transplant. 1995;
disease in adults with acute lymphoblastic allogeneic bone marrow transplantation 15(6):825-828.
leukemia undergoing allogeneic facilitated by the Japan Marrow Donor
hematopoietic stem cell transplantation. Clin Program. Biol Blood Marrow Transplant. 29. Sullivan KM, Shulman HM, Storb R, et al.
Lymphoma Myeloma Leuk. 2014;14(4): 2017;23(6):938-944. Chronic graft-versus-host disease in 52
319-326. patients: adverse natural course and
16. Passweg JR, Tiberghien P, Cahn JY, et al. successful treatment with combination
5. Bar M, Wood BL, Radich JP, et al. Impact of Graft-versus-leukemia effects in T lineage immunosuppression. Blood. 1981;57(2):
minimal residual disease, detected by flow and B lineage acute lymphoblastic leukemia. 267-276.
cytometry, on outcome of myeloablative Bone Marrow Transplant. 1998;21(2):
hematopoietic cell transplantation for acute 153-158. 30. Gray RJ. A class of K-sample tests for com-
lymphoblastic leukemia. Leukemia Res Treat. paring the cumulative incidence of a
2014;2014:421723. 17. Goekbuget N, Dombret H, Bonifacio M, competing risk. Ann Stat. 1988;16(3):
et al. BLAST: a confirmatory, single-arm, 1141-1154.
6. Leung W, Pui CH, Coustan-Smith E, et al. phase 2 tudy of blinatumomab, a Bispecific
Detectable minimal residual disease before T-Cell Engager (BiTEV R ) antibody construct, 31. Cai G, Anumanthan A, Brown JA, Greenfield
hematopoietic cell transplantation is in patients with minimal residual disease EA, Zhu B, Freeman GJ. CD160 inhibits
prognostic but does not preclude cure for B-precursor acute lymphoblastic leukemia activation of human CD41 T cells through
children with very-high-risk leukemia. Blood. (ALL). Blood. 2014;124(21):379-379. interaction with herpesvirus entry mediator.
2012;120(2):468-472. Nat Immunol. 2008;9(2):176-185.
18. Topp MS, G€ okbuget N, Stein AS, et al.
7. Mr
ozek K, Harper DP, Aplan PD. Safety and activity of blinatumomab for 32. Zhang L, Zhang A, Xu J, et al. CD160 plays a
Cytogenetics and molecular genetics of adult patients with relapsed or refractory protective role during chronic infection by
acute lymphoblastic leukemia. Hematol B-precursor acute lymphoblastic leukaemia: enhancing both functionalities and
Oncol Clin North Am. 2009;23(5): a multicentre, single-arm, phase 2 study. proliferative capacity of CD81 T cells. Front
991-1010, v. Lancet Oncol. 2015;16(1):57-66. Immunol. 2020;11:2188.
8. Chennamaneni R, Gundeti S, Konatam ML, 19. von Stackelberg A, Locatelli F, Zugmaier G, 33. Duell J, Dittrich M, Bedke T, et al.
Bala S, Kumar A, Srinivas L. Impact of et al. Phase I/phase II study of Frequency of regulatory T cells determines
cytogenetics on outcomes in pediatric acute blinatumomab in pediatric patients with the outcome of the T-cell-engaging
lymphoblastic leukemia. South Asian relapsed/refractory acute lymphoblastic antibody blinatumomab in patients with
J Cancer. 2018;7(4):263-266. leukemia. J Clin Oncol. 2016;34(36): B-precursor ALL. Leukemia. 2017;31(10):
4381-4389. 2181-2190.
9. Lazaryan A, Dolan M, Zhang MJ, et al; Acute
Leukemia Committee of the CIBMTR. Impact 20. Kantarjian H, Stein A, G€
okbuget N, et al. 34. Puzzolo MC, Radice G, Peragine N, et al.
of cytogenetic abnormalities on outcomes of Blinatumomab versus chemotherapy for Host immune system modulation in Ph1
adult Philadelphia-negative acute lympho- advanced acute lymphoblastic leukemia. acute lymphoblastic leukemia patients
blastic leukemia after allogeneic hematopoi- N Engl J Med. 2017;376(9):836-847. treated with dasatinib and blinatumomab.
etic stem cell transplantation: a study by the
Blood. 2021;138(22):2290-2293.
Acute Leukemia Working Committee of the 21. Stein AS, Kantarjian H, G€okbuget N, et al.
Center for International Blood and Marrow Blinatumomab for acute lymphoblastic 35. Dwarakanath BS, Farooque A, Gupta S.
Transplant Research. Haematologica. 2020; leukemia relapse after allogeneic Targeting regulatory T cells for improving
105(5):1329-1338. hematopoietic stem cell transplantation. Biol cancer therapy: challenges and prospects.
Blood Marrow Transplant. 2019;25(8):
10. Poon LM, Hamdi A, Saliba R, et al. Cancer Rep (Hoboken). 2018;1(1):
1498-1504.
Outcomes of adults with acute e21105.
lymphoblastic leukemia relapsing after 22. Zeiser R, Vago L. Mechanisms of immune
36. Suryadevara CM, Desai R, Farber SH, et al.
allogeneic hematopoietic stem cell escape after allogeneic hematopoietic cell
Preventing Lck activation in CAR T cells
transplantation. Biol Blood Marrow transplantation. Blood. 2019;133(12):
confers Treg resistance but requires 4-1BB
Transplant. 2013;19(7):1059-1064. 1290-1297.
signaling for them to persist and treat solid
11. Poon LM, Bassett R Jr, Rondon G, et al. 23. Amgen Inc. Blincyto (blinatumomab). U.S. tumors in nonlymphodepleted hosts. Clin
Outcomes of second allogeneic Food and Drug Administration website. Cancer Res. 2019;25(1):358-368.
hematopoietic stem cell transplantation for www.accessdata.fda.gov/drugsatfda_docs/
patients with acute lymphoblastic leukemia. 37. Zhu M, Kratzer A, Johnson J, et al.
label/2018/125557s013lbl.pdf. Revised
Bone Marrow Transplant. 2013;48(5): Blinatumomab pharmacodynamics and
March 2018. Accessed 14 September 2021.
666-670. exposure-response relationships in relapsed/
24. Amir AD, Davis KL, Tadmor MD, et al. viSNE refractory acute lymphoblastic leukemia. J
12. Bader P, Kreyenberg H, Hoelle W, et al. enables visualization of high dimensional Clin Pharmacol. 2018;58(2):
Increasing mixed chimerism is an important single-cell data and reveals phenotypic het- 168-179.
prognostic factor for unfavorable outcome in erogeneity of leukemia. Nat Biotechnol.
children with acute lymphoblastic leukemia 2013;31(6):545-552. 38. Ueda M, de Lima M, Caimi P, et al.
after allogeneic stem-cell transplantation: Concurrent blinatumomab and donor
possible role for pre-emptive immunother- 25. van der Maaten L. Accelerating t-SNE using lymphocyte infusions for treatment of
apy? J Clin Oncol. 2004;22(9):1696-1705. tree-based algorithms. J Mach Learn. 2014; relapsed pre-B-cell ALL after allogeneic
15:3221-3245. hematopoietic cell transplant. Bone Marrow
13. Collins RH Jr, Shpilberg O, Drobyski WR, Transplant. 2016;51(9):1253-1255.
et al. Donor leukocyte infusions in 140 26. Levine JH, Simonds EF, Bendall SC, et al.
patients with relapsed malignancy after Data-driven phenotypic dissection of AML 39. Mackall CL, Fry TJ, Gress RE. Harnessing the
allogeneic bone marrow transplantation. reveals progenitor-like cells that correlate biology of IL-7 for therapeutic application.
J Clin Oncol. 1997;15(2):433-444. with prognosis. Cell. 2015;162(1):184-197. Nat Rev Immunol. 2011;11(5):330-342.
14. Sullivan KM, Storb R, Buckner CD, et al. 27. CIBMTR. Pre-HCT preparative regimen (con- 40. Perales MA, Goldberg JD, Yuan J, et al.
Graft-versus-host disease as adoptive ditioning). 2020: Available at: https://www. Recombinant human interleukin-7 (CYT107)
immunotherapy in patients with advanced cibmtr.org/manuals/fim/1/en/topic/q155- promotes T-cell recovery after allogeneic
BLINATUMOMAB IN B-ALL POST–ALLOGENEIC HCT blood® 24 MARCH 2022 | VOLUME 139, NUMBER 12 1919