Novel Immunotherapeutics in B-Cell Precursor ALL
Novel Immunotherapeutics in B-Cell Precursor ALL
Review
Optimal Use of Novel Immunotherapeutics in B-Cell
Precursor ALL
Federico Lussana 1,2, * , Gianluca Cavallaro 1,2 , Pantaleo De Simone 1,2 and Alessandro Rambaldi 1,2
Simple Summary: In this review, we discussed the impact of novel immune therapies on the
treatment of child and adult acute lymphoblastic leukemia (ALL). Based on the promising results
achieved in the relapsed/refractory (R/R) setting, several trials are currently evaluating the clinical
benefit of incorporating these new drugs into frontline treatments. The results emerging from the
most recent studies are opening new avenues to further the significant improvements in the clinical
outcome of this disease.
Abstract: Novel immune therapies are currently being used for patients with R/R ALL based on
their ability to induce not only hematologic but also molecular remission. Despite promising results,
specific clinical conditions, such as high tumor burden or extra medullary relapse, are still associated
with a remarkably poor clinical outcome. Therefore, how to optimize the choice and the timing of such
new treatments within different clinical settings remains a matter of debate. In addition, with the aim
of increasing the rate and depth of molecular remission, clinical studies are currently evaluating the
combination of these immunotherapies with chemotherapy in the contest of frontline treatment. The
preliminary data suggest that this approach may increase the cure rate and perhaps reduce the use of
allogeneic stem cell transplantation (alloHSCT) in first remission. In Ph-positive ALL, reproducible
results are showing that frontline treatment programs, based on the combination of tyrosine kinase
inhibitors and immunotherapy, can achieve unprecedented rates of hematologic and molecular
remission as well as a long-term cure, even in the absence of chemotherapy and alloHSCT. The results
Citation: Lussana, F.; Cavallaro, G.; from these studies have led to the development of potentially curative treatment modalities, even for
De Simone, P.; Rambaldi, A. Optimal
older ALL patients who cannot be treated with conventional intensive chemotherapy. The present
Use of Novel Immunotherapeutics in
review examined the evidence for an appropriate use of the new immunotherapies in ALL patients
B-Cell Precursor ALL. Cancers 2023,
and provided some appraisal of the current and future possible uses of these drugs for achieving
15, 1349. https://doi.org/10.3390/
further therapeutic improvement in the treatment of this disease.
cancers15041349
Academic Editors: Elisabetta Keywords: B-cell precursor acute lymphoblastic leukemia; relapsed/refractory; blinatumomab;
Abruzzese and William Arcese inotuzumab; CAR-T cells
Received: 6 January 2023
Revised: 5 February 2023
Accepted: 15 February 2023
Published: 20 February 2023 1. Introduction
Acute lymphoblastic leukemia (ALL) is a hematologic malignancy that originates
from the proliferation of a single B- or T-lymphocyte progenitor, which proliferates and
accumulates in the blood; the bone marrow; and, possibly, the extramedullary sites. This
Copyright: © 2023 by the authors.
disease represents the most frequent cancer in the pediatric population, especially in infants
Licensee MDPI, Basel, Switzerland.
aged 1–4 years, with a subsequent decrease in the overall incidence which reaches its lowest
This article is an open access article
point in young adults aged 25–45 years. The majority of ALL is of B-cell origin. Remarkable
distributed under the terms and
advances have been achieved in the treatment of ALL thanks to the optimal use of intensive
conditions of the Creative Commons
chemotherapy, the implementation of minimal residual disease monitoring, and better sup-
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
portive care, allowing most pediatric and adult patients to achieve a complete hematologic
4.0/).
remission (CR). Despite these significant advances in treatment, less than 50% of adult
patients survive 5 years or more, and about 15–20% of children suffer a relapse. Relapsed
or refractory (R/R) ALL is still a major concern, with a dismal prognosis in both children
and adults and with an overall survival of 3 to 12 months depending on the duration of
the first remission and the number of lines of salvage therapy. In the last few years, the
advent of novel targeted immunotherapies, specifically blinatumomab, inotuzumab, and
chimeric antigen receptor T cells (CAR-T cells), has changed the therapeutic landscape of
R/R B-cell precursor ALL (BCP-ALL). These novel immunotherapies showed impressive
results in terms of response rates and a greater likelihood of proceeding to allogeneic stem
cell transplantation (alloHSCT) consolidation, thus significantly changing the perspectives
of this very unfavorable condition. Based on the promising results in the R/R setting,
immunotherapies are currently under investigation in the early phases of the disease. More-
over, these targeted immunotherapies are usually well tolerated, so the associated clinical
benefit is not limited to younger and fit patients but also to older and frail patients. The
main objectives of this review were to focus on how to choose among these novel agents
according to specific conditions and to highlight the important challenges to be addressed
in the coming years.
2. Immunotherapies
2.1. Blinatumomab
Bispecific T-cell engagers (BiTE) are a class of bispecific antibodies designed for cancer
immunotherapy [1]. Among these, blinatumomab is a bispecific T-cell engager with a
dual affinity for CD3 and CD19 that brings effector T lymphocytes and target B cells into
close proximity, allowing T cells to recognize and exert their cytotoxic activity against
CD19-positive B cells. Blinatumomab has been approved by the US Food and Drug Ad-
ministration for the treatment of pediatric and adult patients with R/R BCP-ALL as well
as for the treatment of those with measurable residual disease (MRD). Based on the re-
sults of the phase II studies on R/R adult ALL [2,3], the current treatment schedule is
based on a ramp-up with 9 mcg daily over the first week followed by 28 mcg daily for
28 days in each subsequent cycle. The efficacy and safety of blinatumomab for the treat-
ment of adult patients with R/R Ph-negative B-cell precursor ALL have been demonstrated
through the pivotal, phase III TOWER study (ClinicalTrials.gov identifier: NCT02013167)
that randomized (2:1 ratio) 405 patients to either blinatumomab monotherapy or multia-
gent chemotherapy [4]. Compared with the chemotherapy arm, the blinatumomab-treated
patients achieved a significantly better complete remission (CR, 34% versus 16%), event-
free survival (EFS, 31% versus 12%), and median overall survival (OS, 7.7 months versus
4.4 months), no matter their age, prior to salvage treatment or prior to alloHSCT. Among
the patients who achieved CR, 76% were MRD negative in the blinatumomab treatment
group versus 48% in the chemotherapy group. These results have been confirmed in
the pediatric setting. In a phase I/II dose-escalation/dose-expansion trial conducted on
Ph-negative ALL patients with a refractory disease or a disease relapsed after at least two
lines of therapy or alloHSCT, blinatumomab showed a complete remission rate of 39%,
with MRD negativity in 52% of the responders. The median OS was 7.5 months [5]. In a
phase III randomized study AALL1331 trial (ClinicalTrials.gov identifier: NCT02101853),
the effect of postreinduction/consolidation with blinatumomab vs. chemotherapy was
evaluated in patients < 30 years. Blinatumomab proved superior compared to chemother-
apy, with a 2-year OS rate of 71% vs. 58%. The rate of MRD negativity was 75% for the
blinatumomab group vs. 32% for the chemotherapy group (p < 0.001). Given the better
response, the likelihood of proceeding alloHSCT was greater for the blinatumomab group
(70% vs. 43%) [6]. In a parallel study, blinatumomab, compared with intensive multidrug
chemotherapy, before alloHSCT was confirmed to have a clinical benefit, with an improved
EFS (66% vs. 27%) and OS (85% vs. 70%) [7].
The efficacy of blinatumomab has also been tested as a single agent in patients with
R/R Ph+ ALL (ALCANTARA; ClinicalTrials.gov identifier: NCT02000427), showing a
response rate of 36% associated with a rate of complete MRD response of 88%, which was
Cancers 2023, 15, 1349 3 of 23
close to that reported in R/R Ph-negative ALL. Among the responders, 44% proceeded to
alloHSCT. After a median follow-up of 16.1 months, the median relapse-free survival (RFS)
was 6.8 months, while the median OS was 9.0 months in the whole cohort and 19.8 months
in the blinatumomab responders [8]. A propensity score analysis was also performed to
compare the outcomes of the patients treated with blinatumomab in the AZCANTARA
study with those of an external historical cohort of patients receiving standard chemother-
apy. A higher rate of CR/CRh (36% vs. 25%) and a better OS with a hazard ratio of 0.81
(95%CI, 0.57 to 1.14) was seen after treatment with blinatumomab [9].
Based on the significant results obtained in the R/R setting, blinatumomab was
also evaluated in patients with molecular evidence of MRD in their first or later CR. In
a multicenter phase II study, which was conducted in 116 adult patients with ALL in
hematologic CR but with a high MRD level (>10−3 ), blinatumomab induced an excellent
78% molecular response rate after the first cycle. The median OS was 36.5 months, and a
prolonged survival was occasionally achieved without alloHSCT [10]. Interestingly, for the
patients in first CR achieving MRD negativity, there was no evidence that the OS improved
in patients who did or did not undergo transplantation. On the contrary, the outcomes
of the patients receiving blinatumomab who were in their second or later CR and who
did not proceed to HCT were inferior [10]. On the basis of these results, the US Food
and Drug Administration (FDA) and the European Medicines Agency (EMA) granted the
first approval for the treatment of MRD with blinatumomab. In a large real-world study,
blinatumomab confirmed strong efficacy outcomes in adults with MRD+ B-cell precursor
ALL, with 91% of Ph− and 59% of Ph+ patients achieving an MRD response within two
cycles of blinatumomab. The overall MRD response did not differ by the CR state (CR1 or
CR2) in both the Ph− and the Ph+ subgroups [11].
Finally, an escalation phase Ib study (NCT04521231) is investigating the safety and
efficacy of subcutaneous blinatumomab for the treatment of adults with R/R BCP-ALL.
Blinatumomab given subcutaneously does not only simplify its administration but also
maximizes the pharmacokinetics and dynamics while decreasing the patients’ discomfort.
Preliminary results are in keeping with this hypothesis [12].
To sum up, despite the high rate of responses leading to the approval of blinatumomab
as a single agent in R/R ALL, the cure rate remains rather limited without subsequent
consolidation with alloHSCT [13]. Based on these observations and on the optimal results
of blinatumomab in the treatment of MRD positive patients, an anticipated use of this drug
in frontline therapy seems to be the best way to optimize its clinical benefit as we discuss
later in this review.
Nonetheless, the survival with the inotuzumab single agent remained suboptimal. For
this reason, inotuzumab was combined with low-intensity chemotherapy based on 8 cycles
of mini-hyper-CVD with or without blinatumomab in 96 patients with BCP-ALL in their
first relapse (29 with blinatumomab and 67 without). The majority of the patients achieved
CR or CRi (80% for the entire cohort and 91% for those in their first relapse). Among
75 evaluable patients, for MRD assessment through MFC, 62 patients (83%) achieved nega-
tive MRD, with higher rates of MRD negativity in those in their first relapse (89%). Forty-
four patients (46%) received alloHSCT. The 3-year OS rate was 33% for the entire cohort
and 42% for those in their first relapse, which was superior to that observed with either ino-
tuzumab or blinatumomab single agents when using a propensity score analysis [16–19]. In
the SWOG1312 phase I trial, inotuzumab was combined with reduced-intensity chemother-
apy with encouraging results. The last reported data showed a composite rate of remission
of 61% among 23 heavily pretreated patients. The treatment was well tolerated, with three
patients experiencing hepatic VOD after their second transplantation or when receiving
the higher dose of inotuzumab [20]. Recently, the single-arm phase II trial AALL1621
(ClinicalTrials.gov identifier: NCT02981628), conducted by the Children’s Oncology Group,
showed that inotuzumab is effective and well tolerated in heavily pretreated children and
adolescents (age of 1–21 years) with R/R CD22-positive BCP-ALL. VOD after hematopoietic
stem-cell transplantation and prolonged cytopenias were notable toxicities, while partial
CD22 expression and lower CD22 site density were associated with a lower likelihood of
response [21].
In summary, inotuzumab is approved as a single agent for relapsed/refractory B-cell
precursor ALL due to the high rate of response in terms of CR and MRD negativity, but
its efficacy remains limited by a short duration of response. As mentioned later in the
review, to improve the clinical benefit of inotuzumab, ongoing studies are exploring its
use in frontline therapies or as a bridge to allogeneic transplants or CAR-T cell therapy.
Finally, VOD is still a concern, but the hematologic community is progressively learning to
prevent this serious complication by avoiding a dual-alkylator conditioning regimen and
by increasing the time between the last dose of inotuzumab and the conditioning regimen.
In the adult patients, although the impressive rate of CR was confirmed, the long-term
outcomes achieved with academic CAR-T cells were initially less significant, with a median
EFS and OS of 6.1 months and 12.9 months [27]. The largest set of safety and efficacy data
on Tisa-cel in the real-world setting included 255 children and young adult ALL patients.
With a median follow-up of 13.4 months and a CR rate of 85.5%, the 12-month duration of
response (DOR), EFS, and OS rates were 60.9%, 52.4%, and 77.2%, respectively [28].
More recently, the single-arm ZUMA-3 trial evaluated KTE-X19 CAR-T cell therapy in
the largest population of adults with R/R B-cell precursor ALL to date. KTE-X19 allowed
39 patients (71%) to achieve CR or CRi, and 97% of these responders obtained MRD
negativity. After a median follow-up of 16 months, the median overall survival was
18 months across all the treated patients. The safety profile of KTE-X19 was manageable
and was without deaths due to cytokine release syndrome. These survival outcomes com-
pare favorably with those achieved by blinatumomab and inotuzumab, leading the FDA
to approve KTE-X19 as the first CAR-T cell product for adult ALL patients. [29]. Overall,
however, a very recent systematic review of 16 studies involving 489 adults confirmed
a very high early remission rate; however, the duration of response remains challeng-
ing [30], and the need for a subsequent allogeneic transplantation is often considered to be
necessary [31–34].
A novel second-generation CD19 CAR-T cell therapy with a fast binding off rate for
CD19 (AUTO1), designed with the aim to reduce toxicity and to improve persistence,
was tested in a phase I study of 20 adult patients with R/R ALL. Interestingly, the study
design planned a double infusion of CAR-T cells at different dosages according to the blast
percentage. The preliminary results of this study were encouraging, with a high remission
rate (85% had a MRD negative response) and an EFS and OS at 12 months of 48% and
64%, respectively. No patients experienced ≥ grade III CRS, and 15% experienced grade III
neurotoxicity that resolved within 72 h with steroids [35]. To improve the clinical results
and to shorten the time from cell collection to infusion into the patient, new laboratory
approaches have been developed by our Chinese colleagues, such as the so-called FasT
CAR-T (F-CAR-T) cell next-day manufacturing platform. The underlying idea is to freeze
the cells a few hours after the viral transduction and to allow CAR-T cell expansion in vivo
after the cells are thawed and infused back into the patient. Reducing the duration of
the ex vivo culture should limit the differentiation of T cells, favoring the preservation
of naïve and stem cell memory T cells in the final product. This is expected to be useful
for obtaining a longer CAR-T cell persistence and, consequently, higher response rates
and a longer duration of response. The preclinical studies and the first human clinical
studies confirmed a superior in vivo CAR-T cell proliferation and expansion compared to
conventional CAR-T cells, which showed a younger phenotype [36]. A similar clinical trial
is also currently ongoing in the US and in Europe with a novel autologous CD19-targeting
CAR-T cell manufactured using the T-Charge™ Platform (YTB323) for the treatment of
R/R CD19-positive lymphoid neoplasms. The T-Charge™ Platform minimizes the ex
vivo culture time and reduces the manufacturing process time to < 2 days. This novel
platform also preserves naïve/TSCM cells in the final product, leading to a potentially
higher potency and persistence [37] (Clinical Trials.gov identifier: NCT03960840).
New approaches to overcoming the main obstacles of CAR-T cell therapy, such as
the high costs and logistical complexity of the transduction viral process, and to allowing
lymphodepleted patients to access CAR-T cell treatment are underway. In addition, the
time of production may be clinically relevant, as some patients may face disease progression
during this time, precluding the treatment itself. In this regard, to replace patient-derived
CAR-T cells with ready-to-go cellular products, such as off-the-shelf allogeneic CAR-T
cells, represents an ambitious goal of the ongoing research. However, the possibility of life-
threatening graft-versus-host disease (GvHD) with an allogenic cell therapy product raises
major concerns. Therefore, clinical research with immune cells other than “conventional” T
cells is underway (Figure 1A,B).
Cancers 2023, 15, 1349 6 of 25
CAR-T cells, represents an ambitious goal of the ongoing research. However, the possibil-
ity of life-threatening graft-versus-host disease (GvHD) with an allogenic cell therapy
Cancers 2023, 15, 1349 6 of 23
product raises major concerns. Therefore, clinical research with immune cells other than
“conventional” T cells is underway (Figure 1A,B).
ALL
ALL represents
representsacute
acutelymphoblastic
lymphoblasticleukemia,
leukemia,CAR
CAR represents chimeric
represents antigen
chimeric recep-
antigen re-
tor, CIK represents cytokine-induced killer cells, CRS represents cytokine release syndrome,
ceptor, CIK represents cytokine-induced killer cells, CRS represents cytokine release syn-
ICANS represents immune-effector-cell-associated neurotoxicity syndrome, NK represents
drome, ICANS represents immune-effector-cell-associated neurotoxicity syndrome, NK
natural killer, GVHD represents graft-versus-host disease, PD represents programmed
represents natural killer, GVHD represents graft-versus-host disease, PD represents pro-
death, T-reg represents regulatory T cells, and TRAC represents T-cell receptor alpha
grammed death, T-reg represents regulatory T cells, and TRAC represents T-cell receptor
constant gene.
alpha constant gene.
The figure was created with BioRender.com.
The figure was created with BioRender.com.
In a recent phase I and II clinical trial (NCT03389035), we examined the treatment
feasibility of BCP-ALL patients who relapsed after alloHSCT with donor-derived anti-
CD19 CAR-T cells (CARCIK-CD19) engineered with the sleeping beauty (SB) transposon.
A complete response was achieved in 18 out of 27 patients (66.7%) and in 16 out of the
21 patients treated with the 2 highest doses (76.2%). A total of 14 (77.8%) of the overall
Cancers 2023, 15, 1349 7 of 23
responders and 13 of the responders at the highest doses (81.3%) were MRD negative. With
a median follow up of 2.8 years, a significantly better EFS was observed in the patients
treated with the highest doses (p = 0.0265). For the 21 patients treated at the 2 highest
dose levels, the median EFS and OS were 4 and 12 months, respectively. The median DOR
of the 16 patients in CR was 9.5 months, with a 6-month DOR of 54.4% (SE = 13.8). CRS
occurred in nine patients (four patients with grade I CRS and five with grade II CRS), and
immune-effector-cell-associated neurotoxicity (ICAN) (grade III) occurred in two patients
at the two highest doses. Although 10 out of 27 had experienced GvHD after the previous
alloHSCT, GvHD never occurred after treatment with CARCIK-CD19 [38,39]. In keeping
with our results, a very recent phase I study of 25 patients treated with allogeneic CAR-
T cells (UCART19), manufactured from healthy donor peripheral blood, confirmed the
safety profile of allogeneic CAR-T cells, with a low risk of inducing GvHD (only 2 patients
developed grade I acute cutaneous GvHD) [40].
Due to their ability to recognize tumor cells in a non-HLA (human leukocyte antigen)-
restricted manner, other “nonconventional” allogeneic cells used in CAR-T cell technology
are natural killer (NK) cells. Preliminary promising results have been reported in different
tumors, such as multiple myeloma or neuroblastoma [41,42], but the observed short-term
responses remain as the main concern for the use of this cell therapy. Liu and colleagues
published a small study including 11 patients with CD19-positive lymphoid tumors. Eight
patients (73%) achieved a hematologic response, and seven (four with lymphoma and
three with CLL) had complete remission. The responses were rapid and were seen within
30 days after infusion at all dose levels. However, most patients had short-term responses,
and five of eight patients needed post-remission therapy [43].
In conclusion, CAR-T cell therapy represents a potential curative treatment for R/R
ALL, particularly when used for a low burden relapse or MRD positive disease. Given
the complexity of this therapy, its possible early placing in ALL therapy remains a major
open spot. Moreover, a subsequent allogeneic transplant is a matter of debate, and some
biological parameters, such as B-cell aplasia and CAR-T cell persistence, could be helpful
parameters in this difficult choice. A longer follow-up on the pivotal clinical trial will
probably address some of these questions. Finally, there are many new platforms developed
with the aim to make manufacturing easier and faster as well as to improve cell therapy
efficacy. In this regard, it is worth noting that the time of production may be clinically
relevant, as some patients may undergo clinical disease progression during this time,
precluding the treatment itself.
A summary of the evidence leading to the regulatory approval of the immunotherapies
(blinatumomab, inotuzumab, tisagenlecleucel, and brexucabtagene autoleucel) for R/R
ALL is presented in Table 1.
Cancers 2023, 15, 1349 8 of 23
Table 1. Summary of the main clinical studies on the immunotherapies approved for treating R/R BCP-ALL (blinatumomab, inotuzumab, tisagenlecleucel, and
brexucabtagene autoleucel).
Trial/Study Setting Study Design Total Patients Immunotherapeutic Treatment Main Results
[Reference]
CR: blinatumomab 34% vs. ChT 16%.
Blinatumomab 9 µg/day first Molecular response: blinatumomab 76% vs.
Tower 2017 [13] R/R Ph-negative adults Phase III 271 blinatumomab vs. 134 ChT
randomized trial week then 28 µg/day c.i. ChT 48%. Median RFS: blinatumomab 7.3
vs. ChT 4.6 months. OS: blinatumomab 7.7
vs. ChT 4.0 months.
Molecular response: blinatumomab 75% vs.
AALL1331 [6]
R/R Ph-negative children and
Phase III randomized trial 107 blinatumomab vs. 109 ChT Blinatumomab 15 µg/m2 day c.i.
young adults (1–30 years) 32% ChT. RFS: blinatumomab 54% vs. ChT
for 28 days 39%. OS: blinatumomab 71% vs. ChT 58%.
54 blinatumomab vs. 54 ChT for Minimal residual disease remission:
Locatelli et al. [7]
High risk first relapse Ph-negative
Phase III randomized trial the 3rd consolidation Blinatumomab 15 µg/m2 day c.i. blinatumomab 90% vs. ChT 54%. EFS:
children before alloHSCT for 28 days blinatumomab 66% vs. ChT 27%. OS:
blinatumomab 85% vs. ChT 70%.
Blinatumomab 9 µg/day first CR: 36%. Molecular response: 88%. Median
ALCANTARA [8] R/R Ph-positive adults Phase II trial 45
week then 28 µg/day c.i. RFS: 6.7 months. OS: 7.1 months.
Blast [10] MRD positive after induction Phase II trial 113 Blinatumomab 15 µg/m2 / Molecular response: 78%. Medina RFS: 18.9
day c.i. months. OS: 36.5 months.
CR: inotuzumab 81% vs. ChT 29%.
Molecular response: inotuzumab 78% vs.
Inotuzumab 0.8 mg/m2 on day 1 ChT 28%. Median RFS: inotuzumab 5
Phase III
INO-VATE ALL [14] R/R Ph-negative or -positive adults randomized trial 109 inotuzumab vs. 109 ChT of each cycle and 0.5 mg/m2 on months vs. ChT 1.8 months. OS:
days 8 and 15 inotuzumab 7.7 vs. ChT 6.7 months. VOD:
inotuzumab 13% vs. ChT 1% (most cases
occurred after alloHSCT).
CR: 58%. MRD response: 67%. Minimal
residual disease measured by flow
Inotuzumab dosing was 0.8 cytometry: 18 (66.7%) had minimal residual
R/R Ph- and Ph+ mg/m2 intravenously on day 1 disease < 0.01%. VOD: 29% among 21
AALL1621 [21] children and adolescents Phase II trial 48
(age of 1–21 years) and 0.5 mg/m2 on days 8 and 15 patients undergoing alloHSCT. Partial
of a 28-day cycle CD22 expression and lower CD22 site
density were associated with a lower
likelihood of response to inotuzumab.
R/R Ph- and Ph+ Single infusion of
children and young adults 79 CR rate: 82%. MRD negativity: 100%.
Eliana study [23–25] Phase II trial 5-year RFS: 49%. 5y OS: 55%.
(age of 3–21) tisagenlecleucel
Figure 2. Mechanisms of relapse after exposure to different immunotherapies. Panel (A) T-cell
Figure 2. Mechanisms of relapse after exposure to different immunotherapies. Panel (A) T-cell
activation and expansion can be impaired by the leukemia-derived mechanisms of immune es-
activation and expansion
cape, primarily antigen losscan be impaired
or phenomena by the
derived leukemia-derived
from mechanisms
T-cell proliferation of antigen
under chronic immune escape,
primarily antigen
stimulation, such asloss orexhaustion.
T-cell phenomena derived
Panel from T-cell
(B) relapses proliferation
after CAR-T under
cell infusion chronic
could antigen stim-
be related
to antigen loss (CD19-relapses) or to impaired CAR-T cell function (CD19+ relapses). Panel
ulation, such as T-cell exhaustion. Panel (B) relapses after CAR-T cell infusion could be related C: re-
sistance to inotuzumab ozogamicin is mainly related to reduced CD22 expression and impaired
to antigen loss (CD19-relapses) or to impaired CAR-T cell function (CD19+ relapses). Panel C: re-
drug internalization.
sistance to inotuzumab ozogamicin is mainly related to reduced CD22 expression and impaired
drug internalization.
Cancers 2023, 15, 1349 11 of 23
Based on theDisease
4.2. High-Burden limited
at experience
Relapse in the previously quoted studies, at our institution
we considered
In both theinotuzumab toassessing
pivotal trials be the first option
the valuefor
of R/R ALL patients
blinatumomab with
and extramedullary
inotuzumab, high-
disease. For patients with limited SNC involvement, CAR-T cells may represent
burden disease was defined by a marrow blast count greater than 50%. The use a valuable
of blina-
option. in this setting was clearly associated with a reduced probability of achieving
tumomab
CR [4,77]. Even in the setting of MRD, the disease burden matters when blinatumomab
4.2.
is High-Burden
employed. Disease
In the at Relapse
recent retrospective analysis of the GRAAL group, the 3-year OS of
In both
patients withthe pivotal trials
molecular MRD assessing
< 0.1%, the valueor
0.1–1%, of >1%
blinatumomab
was 86%, and 58%,inotuzumab,
and 33%, respec- high-
burden disease was defined by a marrow blast count greater than
tively [78]. These results suggest that an optimal ratio between the CD3+ T cells and the 50%. The use of blina-
tumomab
target CD19+in this setting
cells was clearly
is probably a crucialassociated
parameter.with For
a reduced probability
this reason, of achieving
pre-blinatumomab
CR [4,77].
mild Even in the
cytoreduction settingbeofconsidered
should MRD, the diseasewhenever burden matters
possible when blinatumomab
to improve leukemic con- is
employed.
trol and to In the recent
reduce retrospective
the risk of cytokine analysis
releaseofsyndrome
the GRAAL group,
(CRS) andthe 3-year OS
neurologic of pa-
toxicity
(ICANS)
tients with [52,53,79].
molecular MRD < 0.1%, 0.1–1%, or >1% was 86%, 58%, and 33%, respectively
[78]. A postresults
These hoc analysis
suggestofthat
the anINOVATE
optimal study [15] divided
ratio between patients
the CD3+ intoand
T cells three
thegroups
target
according to their baseline blast counts: low (< 50%), moderate (50–90%),
CD19+ cells is probably a crucial parameter. For this reason, pre-blinatumomab mild cy- and high (>90%).
The authors should
toreduction found be thatconsidered
inotuzumab’s activity
whenever is independent
possible to improvefrom the disease
leukemic controlburden;
and to
moreover,
reduce thethe rateofofcytokine
risk MRD negativity
release among
syndrome the responders
(CRS) and did not seemtoxicity
neurologic to be affected
(ICANS) by
the baseline disease burden. In contrast to blinatumomab, a significant clinical benefit was
[52,53,79].
also observed
A post hoc in analysis
the patients
of thewith a high disease
INOVATE studyburden.
[15] divided patients into three groups
Similar to blinatumomab, the impact
according to their baseline blast counts: low (< 50%), of the disease
moderateburden in CAR-T
(50–90%), and highcell therapy
(> 90%).
seems to be unfavorable. In a study from MSKCC [27], patients
The authors found that inotuzumab’s activity is independent from the disease burden; with a higher disease
burden (BM > 5% blasts) showed a lower CR rate. A recent
moreover, the rate of MRD negativity among the responders did not seem to be affectedwork on a large pediatric
population
by the baselineanalyzing
diseasethe impactInofcontrast
burden. blinatumomab and subsequent
to blinatumomab, anti-CD19
a significant CAR-T
clinical cell
benefit
therapy
was alsoshowed
observedthat a nonresponse
in the patients withtoa blinatumomab
high disease burden.and a high burden disease (>5%
blastsSimilar
in the bone marrow) werethe
to blinatumomab, associated
impact ofwith the the worstburden
disease EFS and in RFS
CAR-T independent
cell therapyof
other prognostic factors [80].
seems to be unfavorable. In a study from MSKCC [27], patients with a higher disease bur-
In summary,
den (BM > 5% blasts)these data suggest
showed a loweraCR preference for inotuzumab
rate. A recent work on a largefor patients
pediatric with high-
popula-
burden disease. In such cases, a proper tumor lysis syndrome
tion analyzing the impact of blinatumomab and subsequent anti-CD19 CAR-T cell therapyprophylaxis and, possibly,
inpatient
showed that management
a nonresponse for theto first cycle couldand
blinatumomab be considered.
a high burden Thedisease
use of blinatumomab
(> 5% blasts in
likely remains preferable in patients lacking a robust expression
the bone marrow) were associated with the worst EFS and RFS independent of other of CD22, those whoprog-
were
previously
nostic factors treated
[80]. with inotuzumab, or those with a higher risk of hepatotoxicity. In this
respect, no more than two cycles of inotuzumab should be advised in patients who are
In summary, these data suggest a preference for inotuzumab for patients with high-
candidates for subsequent consolidation with allogeneic transplantation. Inotuzumab may
burden disease. In such cases, a proper tumor lysis syndrome prophylaxis and, possibly,
also represent an ideal bridging therapy for CAR-T cells due to the possibility of providing
inpatient management for the first cycle could be considered. The use of blinatumomab
a sequential target therapy on two different antigens.
likely remains preferable in patients lacking a robust expression of CD22, those who were
Cancers 2023, 15, 1349 13 of 23
4.3. Blinatumomab, Inotuzumab, and CAR-T Cells: Do They Stand Alone or Bridge Treatments to
Allogeneic Transplants?
At present, the need for subsequent alloHSCT in patients successfully treated with
blinatumomab, inotuzumab, or CAR-T cells remains a matter of debate. This uncertainty is
partly due to the fact that none of the published studies were designed to assess the role
of alloHSCT in posttreatment consolidation, and the decision for alloHSCT was left to the
treating physician. With these limitations, a clear clinical benefit for patients proceeding to
alloHSCT after blinatumomab has not yet been demonstrated [3,81] but has been repeatedly
suggested [11,82]. In particular, blinatumomab maintenance therapy (≥ six cycles) was
associated with an increased OS and RFS compared with patients not receiving maintenance
independently from consolidation with alloHSCT [83]. In the MRD setting, the OS was
not different in patients who did or did not undergo transplantation, whereas, for those
treated during their second or later CR, the outcome of patients who did not undergo
transplantation was inferior [10]. Nonetheless, in a long-term analysis of a phase II study
conducted in the MRD setting, younger patients (≤ 35 years) who underwent alloHSCT
had a better 3-year survival compared with those who did not (62% vs. 22%) [82]. Finally, a
single-center experience confirmed alloHSCT after blinatumomab salvage therapy to be an
effective postremission treatment, with a 2-year RFS of 40% [84].
A post hoc analysis of the INOVATE trial revealed better outcomes in the patients
achieving MRD negativity and proceeding to alloHSCT compared to the nontransplanted
subjects [85,86]. In a pooled analysis of the phase I/II and phase III studies, the best
outcome was observed in the patients who underwent alloHSCT directly upon first re-
mission [87]. The median OS was not reached with a 2-year survival probability of 51%.
As expected, the MRD negative patients had a better median posttransplant OS and PFS
compared to the MRD positive patients (17.8 months vs. 5 months and 10.8 months vs.
3.4 months, respectively).
When considering CAR-T cells, different studies showed high rates of response and
a prolonged survival with a different proportion of patients subsequently treated with
alloHSCT after treatment with CAR-T cells. The pivotal phase II trial ELIANA showed
a rate of RFS up to 60% at 1 year and 49% at 5 years, with only a minority of patients
receiving alloHSCT [24,25]. Using a different CAR-T construct, Lee et al. demonstrated
a role of transplantation in consolidation in reducing the relapse rate in young patients
with R/R BCP-ALL [88]. Similarly, in a phase I trial using CD19.28 CAR-T cells in R/R
children and AYAs, the patients with a molecular response who received a transplant
showed a sustained response and survival at long-term follow-up. All the MRD negative
patients not proceeding to alloHSCT relapsed at a median time of 5 months post-CAR-T
cell infusion [76]. In the adult setting, conflicting results have been reported on the role of
consolidative alloHSCT in adult patients achieving MRD negative remission after CAR-T
cell therapy. Park and colleagues showed no differences in the survival outcomes between
the patients receiving alloHSCT after achieving MRD negativity compared to those who
did not [27]. Similarly, when analyzing the recent results of the phase II ZUMA-3 trial, Shah
and colleagues showed no effect of alloHSCT on the duration of response nor on the RFS.
However, the proportion of the patients receiving transplantation was significantly low
(18%) [29]. In contrast, three other studies showed that alloHSCT in adult patients achieving
MRD negativity after CD19 CAR-T cell therapy was associated with an improvement in the
EFS. However, this benefit did not translate into a clear benefit in terms of the OS [31–33].
All in all, with the novel immunotherapies, a prolonged clinical benefit can be achieved
in a fair proportion of the responding patients who also achieved an MRD negative status.
In this context, a subsequent alloHSCT should be taken into consideration as a possible
effective strategy at least for younger and fit patients in order to limit the risk of an early
relapse after immunotherapy.
Table 2 summarizes our suggested approach according to different clinical conditions.
Cancers 2023, 15, 1349 14 of 23
5. Future Perspectives
5.1. Anti-CD19 and Anti-CD22 Immunotherapies as Frontline Treatments
Both anti-CD19 and anti-CD22 immunotherapies have been explored in recent clinical
trials as frontline treatments in association or sequentially to chemotherapy. In the phase II
study GIMEMA LAL2317 (NCT03367299), blinatumomab was added to a pediatric-like
risk-oriented regimen in a sequential manner following the early and late consolidation
phases based on high-dose methotrexate and ara-C. Upfront alloHSCT was limited to
patients with a very high clinical or cytogenetic risk profile at diagnosis and patients re-
maining MRD positive (i.e., ≥ 10−4 ) after early consolidation. A total of 149 BCP-ALL
Ph-negative patients were enrolled (median age of 41 and range of 18–65) of whom 90%
obtained a complete hematological response. No matter the MRD status after consolida-
tion, blinatumomab was given to all the patients. MRD negativity improved from 72%
achieved after consolidation chemotherapy up to 95% following one blinatumomab cycle
(p = 0.018) [89]. The use of blinatumomab in first-line treatment was also recently explored
in a phase II single-arm monocentric study by the MD Anderson researchers using a se-
quential approach of four cycles (instead of eight) of high-dose induction chemotherapy
Cancers 2023, 15, 1349 15 of 23
5.2. Should Less Intensive Chemotherapy or a Chemo-Free Approach Be Used to Treat Old and
Frail Patients?
The treatment landscape of older patients is rapidly changing, and their results are
improving thanks to the addition of innovative immunotherapies. The dismal outcome of
older patients with an expected 5-year overall survival of less than 20% [92] will rapidly
improve thanks to these innovative treatments. The MD Anderson group proposed a
reduced-intensity chemotherapy based on eight cycles of mini-hyper-CVD associated
with fractionated inotuzumab for the first four cycles for newly diagnosed patients aged
≥ 60 years. The study protocol was further amended to incorporate four cycles of blina-
tumomab as a consolidation treatment, reducing the chemotherapy to four cycles and
lowering the dose of fractionated inotuzumab. Blinatumomab was continued as a mainte-
nance treatment alternated with POMP. Among 64 patients evaluable for a morphologic
response with a median age of 68 years (range of 60–81), 96% achieved an MRD negativity
overall. The induction mortality was very low (< 5%), and the incidence of VOD was
< 10% [93]. Interestingly, a recent propensity score analysis on 58 old patients treated with
mini-HCVD and inotuzumab revealed improved complete responses and outcomes when
compared to historic controls [19]. The German cooperative group GMALL evaluated the
efficacy of inotuzumab as an induction treatment followed by a conventional high-dose con-
solidation chemotherapy. Patients aged ≥ 55 years were enrolled in this study. Complete
Cancers 2023, 15, 1349 16 of 23
remission was achieved by all the 27 patients who completed the induction phase, with
74% of the patients available for the MRD study reaching a molecular response. Reported
severe adverse events were mainly hematological, while approximately one-third of the
patients experienced a grade ≥ III transaminitis. No VOD cases were reported [94].
The role of blinatumomab as the first-line treatment for older patients with BCP-ALL
was investigated in the SWOG 1318 trial. In this chemo-free protocol, the bispecific antibody
was given as an induction treatment for two cycles followed by an additional three consoli-
dation cycles and POMP maintenance. Treatment was well tolerated, with no early deaths
reported during the induction phase. Of the 29 evaluable patients (median age of 75 years),
two-thirds obtained a complete response, with an estimated 3-year DFS and OS of 37% and
37%, respectively [95]. In line with previous studies, two phase II clinical trials performed
by the European working group on adult ALL (EWALL) are evaluating the combination of
either inotuzumab (NCT03249870) or blinatumomab (NCT03480438) with mild-intensity
chemotherapy in patients older than 55 years (see the Supplementary Table S1).
6. Conclusions
Based on the results obtained in the R/R setting showing the ability of immunothera-
pies to induce not only hematologic but also molecular remission, several trials are currently
evaluating their use as part of frontline treatments. All these trials may lead to the de-
velopment of an innovative combination of chemoimmunotherapies for patients with
ALL with less toxicity and to the establishment of their best positioning in future upfront
therapeutic regimens with the goal of increasing the rate and quality of deep molecular
responses in ALL (Figure 1A,B and Table S1 in the Supplementary Materials). In our opin-
ion, blinatumomab or inotuzumab could be better incorporated into the earlier phases of
treatment based on some characteristics, such as their availability off the shelf and no need
of lymphodepleting chemotherapy, leaving a wider therapeutic role of CAR-T cells in an
earlier R/R setting. The encouraging preliminary results coming from the ongoing studies
suggest that immunotherapies really have the potential to reduce the need to use intensive
chemotherapy or to reduce the indication of alloHSCT as a consolidation treatment. In
this regard, the combination of blinatumomab and the new generation of TKIs in Ph+
ALL represents the first evidence that a frontline chemotherapy-free regimen can be very
effective, also avoiding the need for alloHSCT in most patients. Thus, the landscape of
treatment is now really changing, and, in the coming years, a wider use of these drugs in
frontline treatments is likely. The clinical benefit of incorporating these drugs will not be
limited to younger and fit patients but will also apply to older and more frail patients when
considering that the immunotherapies are better tolerated than conventional chemotherapy.
At the same time, these immunotherapies could improve transplantation outcomes when
given either as a prophylaxis or as a postrelapse treatment [106].
References
1. Einsele, H.; Borghaei, H.; Orlowski, R.Z.; Subklewe, M.; Roboz, G.J.; Zugmaier, G.; Kufer, P.; Iskander, K.; Kantarjian, H.M. The
BiTE (Bispecific T-Cell Engager) Platform: Development and Future Potential of a Targeted Immuno-Oncology Therapy across
Tumor Types. Cancer 2020, 126, 3192–3201. [CrossRef]
2. Topp, M.S.; Gokbuget, N.; Zugmaier, G.; Klappers, P.; Stelljes, M.; Neumann, S.; Viardot, A.; Marks, R.; Diedrich, H.; Faul, C.; et al.
Phase II Trial of the Anti-CD19 Bispecific T Cell-Engager Blinatumomab Shows Hematologic and Molecular Remissions in
Cancers 2023, 15, 1349 18 of 23
Patients with Relapsed or Refractory B-Precursor Acute Lymphoblastic Leukemia. J. Clin. Oncol. 2014, 32, 4134–4140. [CrossRef]
[PubMed]
3. Topp, M.S.; Gokbuget, N.; Stein, A.S.; Zugmaier, G.; O’Brien, S.; Bargou, R.C.; Dombret, H.; Fielding, A.K.; Heffner, L.;
Larson, R.A.; et al. Safety and Activity of Blinatumomab for Adult Patients with Relapsed or Refractory B-Precursor Acute
Lymphoblastic Leukaemia: A Multicentre, Single-Arm, Phase 2 Study. Lancet Oncol. 2015, 16, 57–66. [CrossRef] [PubMed]
4. Kantarjian, H.; Stein, A.; Gokbuget, N.; Fielding, A.K.; Schuh, A.C.; Ribera, J.-M.; Wei, A.; Dombret, H.; Foa, R.; Bassan, R.; et al.
Blinatumomab versus Chemotherapy for Advanced Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2017, 376, 836–847.
[CrossRef] [PubMed]
5. Gore, L.; Locatelli, F.; Zugmaier, G.; Handgretinger, R.; O’Brien, M.M.; Bader, P.; Bhojwani, D.; Schlegel, P.-G.; Tuglus, C.A.;
von Stackelberg, A. Survival after Blinatumomab Treatment in Pediatric Patients with Relapsed/Refractory B-Cell Precursor
Acute Lymphoblastic Leukemia. Blood Cancer J. 2018, 8, 80. [CrossRef] [PubMed]
6. Brown, P.A.; Ji, L.; Xu, X.; Devidas, M.; Hogan, L.E.; Borowitz, M.J.; Raetz, E.A.; Zugmaier, G.; Sharon, E.; Bernhardt, M.B.; et al.
Effect of Postreinduction Therapy Consolidation with Blinatumomab vs Chemotherapy on Disease-Free Survival in Children,
Adolescents, and Young Adults with First Relapse of B-Cell Acute Lymphoblastic Leukemia: A Randomized Clinical Trial. JAMA
2021, 325, 833–842. [CrossRef] [PubMed]
7. Locatelli, F.; Zugmaier, G.; Rizzari, C.; Morris, J.D.; Gruhn, B.; Klingebiel, T.; Parasole, R.; Linderkamp, C.; Flotho, C.; Petit, A.; et al.
Effect of Blinatumomab vs. Chemotherapy on Event-Free Survival Among Children with High-Risk First-Relapse B-Cell Acute
Lymphoblastic Leukemia: A Randomized Clinical Trial. JAMA 2021, 325, 843–854. [CrossRef]
8. Martinelli, G.; Boissel, N.; Chevallier, P.; Ottmann, O.; Gokbuget, N.; Topp, M.S.; Fielding, A.K.; Rambaldi, A.; Ritchie, E.K.;
Papayannidis, C.; et al. Complete Hematologic and Molecular Response in Adult Patients with Relapsed/Refractory Philadelphia
Chromosome-Positive B-Precursor Acute Lymphoblastic Leukemia Following Treatment with Blinatumomab: Results from a
Phase II, Single-Arm, Multicenter Study. J. Clin. Oncol. 2017, 35, 1795–1802. [CrossRef]
9. Rambaldi, A.; Ribera, J.-M.; Kantarjian, H.M.; Dombret, H.; Ottmann, O.G.; Stein, A.S.; Tuglus, C.A.; Zhao, X.; Kim, C.;
Martinelli, G. Blinatumomab Compared with Standard of Care for the Treatment of Adult Patients with Relapsed/Refractory
Philadelphia Chromosome-Positive B-Precursor Acute Lymphoblastic Leukemia. Cancer 2020, 126, 304–310. [CrossRef]
10. Gokbuget, N.; Dombret, H.; Bonifacio, M.; Reichle, A.; Graux, C.; Faul, C.; Diedrich, H.; Topp, M.S.; Bruggemann, M.;
Horst, H.-A.; et al. Blinatumomab for Minimal Residual Disease in Adults with B-Cell Precursor Acute Lymphoblastic Leukemia.
Blood 2018, 131, 1522–1531. [CrossRef]
11. Boissel, N.; Chiaretti, S.; Papayannidis, C.; Ribera, J.M.; Bassan, R.; Sokolov, A.N.; Alam, N.; Brescianini, A.; Pezzani, I.;
Kreuzbauer, G.; et al. Real-World Use of Blinatumomab in Adult Patients with B-Cell Acute Lymphoblastic Leukemia in Clinical
Practice: Results from the NEUF Study. Blood Cancer J. 2023, 13, 2. [CrossRef] [PubMed]
12. Martínez Sánchez, P.; Zugmaier, G.; Gordon, P.; Jabbour, E.; Rifón Roca, J.J.; Schwartz, S.; Borlenghi, E.; Huguet, F.;
Hernández-Rivas, J.M.; Lussana, F.; et al. Safety and Pharmacokinetics of Subcutaneous Blinatumomab (SC Blinatumomab) for
the Treatment of Adults with Relapsed or Refractory B Cell Precursor Acute Lymphoblastic Leukemia (R/R B-ALL); Results from
a Phase 1b Study. Blood 2022, 140, 6122–6124. [CrossRef]
13. Kantarjian, H.M.; DeAngelo, D.J.; Stelljes, M.; Martinelli, G.; Liedtke, M.; Stock, W.; Gökbuget, N.; O’Brien, S.; Wang, K.;
Wang, T.; et al. Inotuzumab Ozogamicin versus Standard Therapy for Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2016, 375,
740–753. [CrossRef]
14. Kantarjian, H.M.; DeAngelo, D.J.; Stelljes, M.; Liedtke, M.; Stock, W.; Gökbuget, N.; O’Brien, S.M.; Jabbour, E.; Wang, T.;
Liang White, J.; et al. Inotuzumab Ozogamicin versus Standard of Care in Relapsed or Refractory Acute Lymphoblastic Leukemia:
Final Report and Long-Term Survival Follow-up from the Randomized, Phase 3 INO-VATE Study. Cancer 2019, 125, 2474–2487.
[CrossRef] [PubMed]
15. DeAngelo, D.J.; Advani, A.S.; Marks, D.I.; Stelljes, M.; Liedtke, M.; Stock, W.; Gökbuget, N.; Jabbour, E.; Merchant, A.;
Wang, T.; et al. Inotuzumab Ozogamicin for Relapsed/Refractory Acute Lymphoblastic Leukemia: Outcomes by Disease Burden.
Blood Cancer J. 2020, 10, 81. [CrossRef] [PubMed]
16. Sasaki, K.; Kantarjian, H.; Ravandi, F.; Short, N.; Kebriaei, P.; Huang, X.; Jain, N.; Konopleva, M.; Garcia-Manero, G.;
Champlin, R.; et al. Long-Term Follow-up of the Combination of Low-Intensity Chemotherapy Plus Inotuzumab Ozogamicin
with or without Blinatumomab in Patients with Relapsed-Refractory Philadelphia Chromosome-Negative Acute Lymphoblastic
Leukemia: A Phase 2 Trial. Blood 2020, 136, 40–42. [CrossRef]
17. Jabbour, E.; Sasaki, K.; Ravandi, F.; Huang, X.; Short, N.J.; Khouri, M.; Kebriaei, P.; Burger, J.; Khoury, J.; Jorgensen, J.; et al.
Chemoimmunotherapy with Inotuzumab Ozogamicin Combined with Mini-Hyper-CVD, with or without Blinatumomab, Is
Highly Effective in Patients with Philadelphia Chromosome-Negative Acute Lymphoblastic Leukemia in First Salvage. Cancer
2018, 124, 4044–4055. [CrossRef]
18. Jabbour, E.; Sasaki, K.; Short, N.J.; Ravandi, F.; Huang, X.; Khoury, J.D.; Kanagal-Shamanna, R.; Jorgensen, J.; Khouri, I.F.;
Kebriaei, P.; et al. Long-Term Follow-up of Salvage Therapy Using a Combination of Inotuzumab Ozogamicin and Mini-
Hyper-CVD with or without Blinatumomab in Relapsed/Refractory Philadelphia Chromosome-Negative Acute Lymphoblastic
Leukemia. Cancer 2021, 127, 2025–2038. [CrossRef]
19. Jabbour, E.J.; Sasaki, K.; Ravandi, F.; Short, N.J.; Garcia-Manero, G.; Daver, N.; Kadia, T.; Konopleva, M.; Jain, N.; Cortes, J.; et al.
Inotuzumab Ozogamicin in Combination with Low-Intensity Chemotherapy (Mini-HCVD) with or without Blinatumomab versus
Cancers 2023, 15, 1349 19 of 23
Standard Intensive Chemotherapy (HCVAD) as Frontline Therapy for Older Patients with Philadelphia Chromosome-Negative
Acute Lymphoblastic. Cancer 2019, 125, 2579–2586. [CrossRef]
20. Advani, A.S.; Moseley, A.; Liedtke, M.; O’Donnell, M.R.; Aldoss, I.; Mims, M.P.; O’Dwyer, K.M.; Othus, M.; Erba, H.P. SWOG
1312 Final Results: A Phase 1 Trial of Inotuzumab in Combination with CVP (Cyclophosphamide, Vincristine, Prednisone) for
Relapsed/ Refractory CD22+ Acute Leukemia. Blood 2019, 134, 227. [CrossRef]
21. O’Brien, M.M.; Ji, L.; Shah, N.N.; Rheingold, S.R.; Bhojwani, D.; Yuan, C.M.; Xu, X.; Yi, J.S.; Harris, A.C.; Brown, P.A.; et al.
Phase II Trial of Inotuzumab Ozogamicin in Children and Adolescents with Relapsed or Refractory B-Cell Acute Lymphoblastic
Leukemia: Children’s Oncology Group Protocol AALL1621. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2022, 40, 956–967.
[CrossRef] [PubMed]
22. Maude, S.L.; Teachey, D.T.; Porter, D.L.; Grupp, S.A. CD19-Targeted Chimeric Antigen Receptor T-Cell Therapy for Acute
Lymphoblastic Leukemia. Blood 2015, 125, 4017–4023. [CrossRef] [PubMed]
23. Maude, S.L.; Frey, N.; Shaw, P.A.; Aplenc, R.; Barrett, D.M.; Bunin, N.J.; Chew, A.; Gonzalez, V.E.; Zheng, Z.; Lacey, S.F.; et al.
Chimeric Antigen Receptor T Cells for Sustained Remissions in Leukemia. N. Engl. J. Med. 2014, 371, 1507–1517. [CrossRef]
[PubMed]
24. Maude, S.L.; Laetsch, T.W.; Buechner, J.; Rives, S.; Boyer, M.; Bittencourt, H.; Bader, P.; Verneris, M.R.; Stefanski, H.E.;
Myers, G.D.; et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N. Engl. J. Med.
2018, 378, 439–448. [CrossRef]
25. Rives, S.; Maude, S.L.; Hiramatsu, H.; Baruchel, A.; Bader, P.; Bittencourt, H.; Buechner, J.; Laetsch, T.; De Moerloose, B.;
Qayed, M.; et al. S112: Tisagenlecleucel In Pediatric and Young Adult Patients (Pts) With Relapsed/Refractory (R/R) B-Cell Acute
Lymphoblastic Leukemia (B-All): Final Analyses from The Eliana Study. HemaSphere 2022, 6, 13–14. [CrossRef]
26. Schultz, L.M.; Baggott, C.; Prabhu, S.; Pacenta, H.L.; Phillips, C.L.; Rossoff, J.; Stefanski, H.E.; Talano, J.-A.; Moskop, A.;
Margossian, S.P.; et al. Disease Burden Affects Outcomes in Pediatric and Young Adult B-Cell Lymphoblastic Leukemia After
Commercial Tisagenlecleucel: A Pediatric Real-World Chimeric Antigen Receptor Consortium Report. J. Clin. Oncol. Off. J. Am.
Soc. Clin. Oncol. 2022, 40, 945–955. [CrossRef]
27. Park, J.H.; Rivière, I.; Gonen, M.; Wang, X.; Sénéchal, B.; Curran, K.J.; Sauter, C.; Wang, Y.; Santomasso, B.; Mead, E.; et al.
Long-Term Follow-up of CD19 CAR Therapy in Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 449–459. [CrossRef]
28. Pasquini, M.C.; Hu, Z.-H.; Curran, K.; Laetsch, T.; Locke, F.; Rouce, R.; Pulsipher, M.A.; Phillips, C.L.; Keating, A.;
Frigault, M.J.; et al. Real-World Evidence of Tisagenlecleucel for Pediatric Acute Lymphoblastic Leukemia and Non-Hodgkin
Lymphoma. Blood Adv. 2020, 4, 5414–5424. [CrossRef]
29. Shah, B.D.; Ghobadi, A.; Oluwole, O.O.; Logan, A.C.; Boissel, N.; Cassaday, R.D.; Leguay, T.; Bishop, M.R.; Topp, M.S.;
Tzachanis, D.; et al. KTE-X19 for Relapsed or Refractory Adult B-Cell Acute Lymphoblastic Leukaemia: Phase 2 Results of the
Single-Arm, Open-Label, Multicentre ZUMA-3 Study. Lancet 2021, 398, 491–502. [CrossRef]
30. Grover, P.; Veilleux, O.; Tian, L.; Sun, R.; Previtera, M.; Curran, E.; Muffly, L. Chimeric Antigen Receptor T-Cell Therapy in Adults
with B-Cell Acute Lymphoblastic Leukemia. Blood Adv. 2022, 6, 1608–1618. [CrossRef]
31. Hay, K.A.; Gauthier, J.; Hirayama, A.V.; Voutsinas, J.M.; Wu, Q.; Li, D.; Gooley, T.A.; Cherian, S.; Chen, X.; Pender, B.S.; et al.
Factors Associated with Durable EFS in Adult B-Cell ALL Patients Achieving MRD-Negative CR after CD19 CAR T-Cell Therapy.
Blood 2019, 133, 1652–1663. [CrossRef]
32. Frey, N.V.; Shaw, P.A.; Hexner, E.O.; Pequignot, E.; Gill, S.; Luger, S.M.; Mangan, J.K.; Loren, A.W.; Perl, A.E.; Maude, S.L.; et al.
Optimizing Chimeric Antigen Receptor T-Cell Therapy for Adults with Acute Lymphoblastic Leukemia. J. Clin. Oncol. Off. J. Am.
Soc. Clin. Oncol. 2020, 38, 415–422. [CrossRef]
33. Jiang, H.; Li, C.; Yin, P.; Guo, T.; Liu, L.; Xia, L.; Wu, Y.; Zhou, F.; Ai, L.; Shi, W.; et al. Anti-CD19 Chimeric Antigen Receptor-
Modified T-Cell Therapy Bridging to Allogeneic Hematopoietic Stem Cell Transplantation for Relapsed/Refractory B-Cell Acute
Lymphoblastic Leukemia: An Open-Label Pragmatic Clinical Trial. Am. J. Hematol. 2019, 94, 1113–1122. [CrossRef]
34. Zhang, X.; Lu, X.; Yang, J.; Zhang, G.; Li, J.; Song, L.; Su, Y.; Shi, Y.; Zhang, M.; He, J.; et al. Efficacy and Safety of Anti-CD19
CAR T-Cell Therapy in 110 Patients with B-Cell Acute Lymphoblastic Leukemia with High-Risk Features. Blood Adv. 2020, 4,
2325–2338. [CrossRef]
35. Roddie, C.; Dias, J.; O’Reilly, M.A.; Abbasian, M.; Cadinanos-Garai, A.; Vispute, K.; Bosshard-Carter, L.; Mitsikakou, M.; Mehra, V.;
Roddy, H.; et al. Durable Responses and Low Toxicity After Fast Off-Rate CD19 Chimeric Antigen Receptor-T Therapy in Adults
with Relapsed or Refractory B-Cell Acute Lymphoblastic Leukemia. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2021, 39, 3352–3363.
[CrossRef]
36. Yang, J.; He, J.; Zhang, X.; Li, J.; Wang, Z.; Zhang, Y.; Qiu, L.; Wu, Q.; Sun, Z.; Ye, X.; et al. Next-Day Manufacture of a Novel
Anti-CD19 CAR-T Therapy for B-Cell Acute Lymphoblastic Leukemia: First-in-Human Clinical Study. Blood Cancer J. 2022,
12, 104. [CrossRef] [PubMed]
37. Flinn, I.W.; Jaeger, U.; Shah, N.N.; Blaise, D.; Briones, J.; Shune, L.; Boissel, N.; Bondanza, A.; Lu, D.; Zhu, X.; et al. A First-in-
Human Study of YTB323, a Novel, Autologous CD19-Directed CAR-T Cell Therapy Manufactured Using the Novel T-Charge TM
Platform, for the Treatment of Patients (Pts) with Relapsed/Refractory (r/r) Diffuse Large B-Cell Lymphoma (DLBCL). Blood
2021, 138, 740. [CrossRef]
Cancers 2023, 15, 1349 20 of 23
38. Magnani, C.F.; Gaipa, G.; Lussana, F.; Belotti, D.; Gritti, G.; Napolitano, S.; Matera, G.; Cabiati, B.; Buracchi, C.; Borleri, G.; et al.
Sleeping Beauty-Engineered CAR T Cells Achieve Antileukemic Activity without Severe Toxicities. J. Clin. Investig. 2020, 130,
6021–6033. [CrossRef]
39. Lussana, F.; Magnani, C.F.; Gaipa, G.; Galimberti, S.; Gritti, G.; Belotti, D.; Napolitano, S.; Buracchi, C.; Borleri, G.M.;
Rambaldi, B.; et al. Final Results of Phase I/II Study of Donor-Derived CAR T Cells Engineered with Sleeping Beauty in Pediatric
and Adult Patients with B-Cell Acute Lymphoblastic Leukemia Relapsed Post-HSCT. Blood 2022, 140, 4568–4569. [CrossRef]
40. Benjamin, R.; Jain, N.; Maus, M.V.; Boissel, N.; Graham, C.; Jozwik, A.; Yallop, D.; Konopleva, M.; Frigault, M.J.; Teshima, T.; et al.
UCART19, a First-in-Class Allogeneic Anti-CD19 Chimeric Antigen Receptor T-Cell Therapy for Adults with Relapsed or
Refractory B-Cell Acute Lymphoblastic Leukaemia (CALM): A Phase 1, Dose-Escalation Trial. Lancet Haematol. 2022, 9, e833–e843.
[CrossRef]
41. Xu, X.; Huang, W.; Heczey, A.; Liu, D.; Guo, L.; Wood, M.; Jin, J.; Courtney, A.N.; Liu, B.; Di Pierro, E.J.; et al. NKT Cells
Coexpressing a GD2-Specific Chimeric Antigen Receptor and IL15 Show Enhanced In Vivo Persistence and Antitumor Activity
against Neuroblastoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 7126–7138. [CrossRef] [PubMed]
42. Poels, R.; Drent, E.; Lameris, R.; Katsarou, A.; Themeli, M.; van der Vliet, H.J.; de Gruijl, T.D.; van de Donk, N.W.C.J.; Mutis, T.
Preclinical Evaluation of Invariant Natural Killer T Cells Modified with CD38 or BCMA Chimeric Antigen Receptors for Multiple
Myeloma. Int. J. Mol. Sci. 2021, 22, 1096. [CrossRef] [PubMed]
43. Liu, E.; Marin, D.; Banerjee, P.; Macapinlac, H.A.; Thompson, P.; Basar, R.; Nassif Kerbauy, L.; Overman, B.; Thall, P.;
Kaplan, M.; et al. Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. N. Engl. J. Med. 2020,
382, 545–553. [CrossRef] [PubMed]
44. Schultz, L.; Gardner, R. Mechanisms of and Approaches to Overcoming Resistance to Immunotherapy. Hematol. Am. Soc. Hematol.
Educ. Progr. 2019, 2019, 226–232. [CrossRef] [PubMed]
45. Aldoss, I.; Forman, S.J. How I Treat Adults with Advanced Acute Lymphoblastic Leukemia Eligible for CD19-Targeted Im-
munotherapy. Blood 2020, 135, 804–813. [CrossRef] [PubMed]
46. Braig, F.; Brandt, A.; Goebeler, M.; Tony, H.-P.; Kurze, A.-K.; Nollau, P.; Bumm, T.; Bottcher, S.; Bargou, R.C.; Binder, M. Resistance
to Anti-CD19/CD3 BiTE in Acute Lymphoblastic Leukemia May Be Mediated by Disrupted CD19 Membrane Trafficking. Blood
2017, 129, 100–104. [CrossRef]
47. Zhao, Y.; Aldoss, I.; Qu, C.; Crawford, J.C.; Gu, Z.; Allen, E.K.; Zamora, A.E.; Alexander, T.B.; Wang, J.; Goto, H.; et al.
Tumor-Intrinsic and -Extrinsic Determinants of Response to Blinatumomab in Adults with B-ALL. Blood 2021, 137, 471–484.
[CrossRef]
48. Aldoss, I.; Song, J.Y. Extramedullary Relapse of KMT2A(MLL)-Rearranged Acute Lymphoblastic Leukemia with Lineage Switch
Following Blinatumomab. Blood 2018, 131, 2507. [CrossRef]
49. Feucht, J.; Kayser, S.; Gorodezki, D.; Hamieh, M.; Doring, M.; Blaeschke, F.; Schlegel, P.; Bosmuller, H.; Quintanilla-Fend, L.;
Ebinger, M.; et al. T-Cell Responses against CD19+ Pediatric Acute Lymphoblastic Leukemia Mediated by Bispecific T-Cell
Engager (BiTE) Are Regulated Contrarily by PD-L1 and CD80/CD86 on Leukemic Blasts. Oncotarget 2016, 7, 76902–76919.
[CrossRef]
50. Duell, J.; Dittrich, M.; Bedke, T.; Mueller, T.; Eisele, F.; Rosenwald, A.; Rasche, L.; Hartmann, E.; Dandekar, T.; Einsele, H.; et al.
Frequency of Regulatory T Cells Determines the Outcome of the T-Cell-Engaging Antibody Blinatumomab in Patients with
B-Precursor ALL. Leukemia 2017, 31, 2181–2190. [CrossRef]
51. Kohnke, T.; Krupka, C.; Tischer, J.; Knosel, T.; Subklewe, M. Increase of PD-L1 Expressing B-Precursor ALL Cells in a Patient
Resistant to the CD19/CD3-Bispecific T Cell Engager Antibody Blinatumomab. J. Hematol. Oncol. 2015, 8, 111. [CrossRef]
[PubMed]
52. Bonifacio, M.; Papayannidis, C.; Lussana, F.; Fracchiolla, N.; Annunziata, M.; Sica, S.; Delia, M.; Foà, R.; Pizzolo, G.; Chiaretti, S.
Real-World Multicenter Experience in Tumor Debulking Prior to Blinatumomab Administration in Adult Patients with Re-
lapsed/Refractory B-Cell Precursor Acute Lymphoblastic Leukemia. Front. Oncol. 2021, 11, 804714. [CrossRef] [PubMed]
53. Jain, T.; Litzow, M.R. Management of Toxicities Associated with Novel Immunotherapy Agents in Acute Lymphoblastic Leukemia.
Ther. Adv. Hematol. 2020, 11. [CrossRef] [PubMed]
54. Lee, D.W.; Kochenderfer, J.N.; Stetler-Stevenson, M.; Cui, Y.K.; Delbrook, C.; Feldman, S.A.; Fry, T.J.; Orentas, R.; Sabatino, M.;
Shah, N.N.; et al. T Cells Expressing CD19 Chimeric Antigen Receptors for Acute Lymphoblastic Leukaemia in Children and
Young Adults: A Phase 1 Dose-Escalation Trial. Lancet 2015, 385, 517–528. [CrossRef]
55. Lamble, A.; Myers, R.M.; Taraseviciute, A.; John, S.; Yates, B.; Steinberg, S.M.; Sheppard, J.D.; Kovach, A.E.; Wood, B.L.;
Borowitz, M.J.; et al. Preinfusion Factors Impacting Relapse Immunophenotype Following CD19 CAR T Cells. Blood Adv. 2022.
[CrossRef]
56. Orlando, E.J.; Han, X.; Tribouley, C.; Wood, P.A.; Leary, R.J.; Riester, M.; Levine, J.E.; Qayed, M.; Grupp, S.A.; Boyer, M.; et al.
Genetic Mechanisms of Target Antigen Loss in CAR19 Therapy of Acute Lymphoblastic Leukemia. Nat. Med. 2018, 24, 1504–1506.
[CrossRef]
57. Sotillo, E.; Barrett, D.M.; Black, K.L.; Bagashev, A.; Oldridge, D.; Wu, G.; Sussman, R.; Lanauze, C.; Ruella, M.; Gazzara, M.R.; et al.
Convergence of Acquired Mutations and Alternative Splicing of CD19 Enables Resistance to CART-19 Immunotherapy. Cancer
Discov. 2015, 5, 1282–1295. [CrossRef]
Cancers 2023, 15, 1349 21 of 23
58. Bueno, C.; Barrera, S.; Bataller, A.; Ortiz-Maldonado, V.; Elliot, N.; O’Byrne, S.; Wang, G.; Rovira, M.; Gutierrez-Agüera, F.;
Trincado, J.L.; et al. CD34+CD19-CD22+ B-Cell Progenitors May Underlie Phenotypic Escape in Patients Treated with CD19-
Directed Therapies. Blood 2022, 140, 38–44. [CrossRef]
59. Fischer, J.; Paret, C.; El Malki, K.; Alt, F.; Wingerter, A.; Neu, M.A.; Kron, B.; Russo, A.; Lehmann, N.; Roth, L.; et al. CD19 Isoforms
Enabling Resistance to CART-19 Immunotherapy Are Expressed in B-ALL Patients at Initial Diagnosis. J. Immunother. 2017, 40,
187–195. [CrossRef]
60. Ruella, M.; Xu, J.; Barrett, D.M.; Fraietta, J.A.; Reich, T.J.; Ambrose, D.E.; Klichinsky, M.; Shestova, O.; Patel, P.R.;
Kulikovskaya, I.; et al. Induction of Resistance to Chimeric Antigen Receptor T Cell Therapy by Transduction of a Sin-
gle Leukemic B Cell. Nat. Med. 2018, 24, 1499–1503. [CrossRef]
61. Hamieh, M.; Dobrin, A.; Cabriolu, A.; van der Stegen, S.J.C.; Giavridis, T.; Mansilla-Soto, J.; Eyquem, J.; Zhao, Z.; Whitlock, B.M.;
Miele, M.M.; et al. CAR T Cell Trogocytosis and Cooperative Killing Regulate Tumour Antigen Escape. Nature 2019, 568, 112–116.
[CrossRef] [PubMed]
62. Perna, F.; Sadelain, M. Myeloid Leukemia Switch as Immune Escape from CD19 Chimeric Antigen Receptor (CAR) Therapy.
Transl. Cancer Res. 2016, 5, S221–S225. [CrossRef] [PubMed]
63. Jacoby, E.; Nguyen, S.M.; Fountaine, T.J.; Welp, K.; Gryder, B.; Qin, H.; Yang, Y.; Chien, C.D.; Seif, A.E.; Lei, H.; et al. CD19 CAR
Immune Pressure Induces B-Precursor Acute Lymphoblastic Leukaemia Lineage Switch Exposing Inherent Leukaemic Plasticity.
Nat. Commun. 2016, 7, 12320. [CrossRef] [PubMed]
64. Gardner, R.; Wu, D.; Cherian, S.; Fang, M.; Hanafi, L.-A.; Finney, O.; Smithers, H.; Jensen, M.C.; Riddell, S.R.; Maloney, D.G.; et al.
Acquisition of a CD19-Negative Myeloid Phenotype Allows Immune Escape of MLL-Rearranged B-ALL from CD19 CAR-T-Cell
Therapy. Blood 2016, 127, 2406–2410. [CrossRef]
65. Sheykhhasan, M.; Manoochehri, H.; Dama, P. Use of CAR T-Cell for Acute Lymphoblastic Leukemia (ALL) Treatment: A Review
Study. Cancer Gene Ther. 2022, 29, 1080–1096. [CrossRef]
66. Paul, M.R.; Wong, V.; Aristizabal, P.; Kuo, D.J. Treatment of Recurrent Refractory Pediatric Pre-B Acute Lymphoblastic Leukemia
Using Inotuzumab Ozogamicin Monotherapy Resulting in CD22 Antigen Expression Loss as a Mechanism of Therapy Resistance.
J. Pediatr. Hematol. Oncol. 2019, 41, e546–e549. [CrossRef]
67. Reinert, J.; Beitzen-Heineke, A.; Wethmar, K.; Stelljes, M.; Fiedler, W.; Schwartz, S. Loss of CD22 Expression and Expansion of a
CD22(Dim) Subpopulation in Adults with Relapsed/Refractory B-Lymphoblastic Leukaemia after Treatment with Inotuzumab-
Ozogamicin. Ann. Hematol. 2021, 100, 2727–2732. [CrossRef]
68. Kantarjian, H.M.; Stock, W.; Cassaday, R.D.; DeAngelo, D.J.; Jabbour, E.J.; O’Brien, S.M.; Stelljes, M.; Wang, T.; Liau, K.F.;
Nguyen, K.; et al. Comparison of CD22 Expression between Baseline, End of Treatment, and Relapse Among Patients Treated
with Inotuzumab Ozogamicin Who Responded and Subsequently Relapsed in Two Clinical Trials. Blood 2018, 132, 2699. [CrossRef]
69. Kantarjian, H.M.; Stock, W.; Cassaday, R.D.; DeAngelo, D.J.; Jabbour, E.; O’Brien, S.M.; Stelljes, M.; Wang, T.; Paccagnella, M.L.;
Nguyen, K.; et al. Inotuzumab Ozogamicin for Relapsed/Refractory Acute Lymphoblastic Leukemia in the INO-VATE Trial:
CD22 Pharmacodynamics, Efficacy, and Safety by Baseline CD22. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021, 27,
2742–2754. [CrossRef]
70. Zheng, S.; Gillespie, E.; Naqvi, A.S.; Hayer, K.E.; Ang, Z.; Torres-Diz, M.; Quesnel-Vallières, M.; Hottman, D.A.;
Bagashev, A.; Chukinas, J.; et al. Modulation of CD22 Protein Expression in Childhood Leukemia by Pervasive Splicing
Aberrations: Implications for CD22-Directed Immunotherapies. Blood Cancer Discov. 2022, 3, 103–115. [CrossRef]
71. Aldoss, I.; Song, J.; Stiller, T.; Nguyen, T.; Palmer, J.; O’Donnell, M.; Stein, A.S.; Marcucci, G.; Forman, S.; Pullarkat, V. Correlates of
Resistance and Relapse during Blinatumomab Therapy for Relapsed/Refractory Acute Lymphoblastic Leukemia. Am. J. Hematol.
2017, 92, 858–865. [CrossRef] [PubMed]
72. Aldoss, I.; Otoukesh, S.; Zhang, J.; Mokhtari, S.; Ngo, D.; Mojtahedzadeh, M.; Al Malki, M.M.; Salhotra, A.; Ali, H.; Aribi, A.; et al.
Extramedullary Disease Relapse and Progression after Blinatumomab Therapy for Treatment of Acute Lymphoblastic Leukemia.
Cancer 2022, 128, 529–535. [CrossRef] [PubMed]
73. Holland, E.M.; Yates, B.; Ling, A.; Yuan, C.M.; Wang, H.-W.; Stetler-Stevenson, M.; LaLoggia, M.; Molina, J.C.; Lichtenstein, D.A.;
Lee, D.W.; et al. Characterization of Extramedullary Disease in B-ALL and Response to CAR T-Cell Therapy. Blood Adv. 2022, 6,
2167–2182. [CrossRef] [PubMed]
74. Frigault, M.J.; Dietrich, J.; Gallagher, K.; Roschewski, M.; Jordan, J.T.; Forst, D.; Plotkin, S.R.; Cook, D.; Casey, K.S.;
Lindell, K.A.; et al. Safety and Efficacy of Tisagenlecleucel in Primary CNS Lymphoma: A Phase 1/2 Clinical Trial. Blood 2022,
139, 2306–2315. [CrossRef]
75. Qi, Y.; Zhao, M.; Hu, Y.; Wang, Y.; Li, P.; Cao, J.; Shi, M.; Tan, J.; Zhang, M.; Xiao, X.; et al. Efficacy and Safety of CD19-Specific
CAR T Cell–Based Therapy in B-Cell Acute Lymphoblastic Leukemia Patients with CNSL. Blood 2022, 139, 3376–3386. [CrossRef]
76. Shah, N.N.; Lee, D.W.; Yates, B.; Yuan, C.M.; Shalabi, H.; Martin, S.; Wolters, P.L.; Steinberg, S.M.; Baker, E.H.; Delbrook, C.P.; et al.
Long-Term Follow-Up of CD19-CAR T-Cell Therapy in Children and Young Adults With B-ALL. J. Clin. Oncol. 2021, 39, 1650–1659.
[CrossRef]
77. Wei, A.H.; Ribera, J.-M.; Larson, R.A.; Ritchie, D.; Ghobadi, A.; Chen, Y.; Anderson, A.; Dos Santos, C.E.; Franklin, J.; Kantarjian, H.
Biomarkers Associated with Blinatumomab Outcomes in Acute Lymphoblastic Leukemia. Leukemia 2021, 35, 2220–2231. [Cross-
Ref]
Cancers 2023, 15, 1349 22 of 23
78. Cabannes-Hamy, A.; Brissot, E.; Leguay, T.; Huguet, F.; Chevallier, P.; Hunault, M.; Escoffre-Barbe, M.; Cluzeau, T.; Balsat, M.;
Nguyen, S.; et al. High Tumor Burden before Blinatumomab Has a Negative Impact on the Outcome of Adult Patients with B-Cell
Precursor Acute Lymphoblastic Leukemia. A Real-World Study by the GRAALL. Haematologica 2022, 107, 2072–2080. [CrossRef]
79. King, A.C.; Bolanos, R.; Velasco, K.; Tu, H.; Zaman, F.; Geyer, M.B.; Park, J.H. Real World Chart Review of Blinatumomab to
Treat Patients with High Disease Burden of Relapsed or Refractory B-Cell Precursor Acute Lymphoblastic Leukemia. Blood 2019,
134, 5079. [CrossRef]
80. Myers, R.M.; Taraseviciute, A.; Steinberg, S.M.; Lamble, A.J.; Sheppard, J.; Yates, B.; Kovach, A.E.; Wood, B.; Borowitz, M.J.;
Stetler-Stevenson, M.; et al. Blinatumomab Nonresponse and High-Disease Burden Are Associated with Inferior Outcomes After
CD19-CAR for B-ALL. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2022, 40, 932–944. [CrossRef]
81. Jabbour, E.J.; Gokbuget, N.; Kantarjian, H.M.; Thomas, X.; Larson, R.A.; Yoon, S.-S.; Ghobadi, A.; Topp, M.S.; Tran, Q.;
Franklin, J.L.; et al. Transplantation in Adults with Relapsed/Refractory Acute Lymphoblastic Leukemia Who Are Treated with
Blinatumomab from a Phase 3 Study. Cancer 2019, 125, 4181–4192. [CrossRef] [PubMed]
82. Topp, M.; Stein, A.S.; Zugmaier, G.; Dombret, H.; Brueggemann, M.; Bonifacio, M.; Dong, X.; Kantarjian, H.M.; Bargou, R.C.;
Gökbuget, N. Long-Term Survival of Adults with B-Cell Precursor (BCP) Acute Lymphoblastic Leukemia (ALL) after Treatment
with Blinatumomab and Subsequent Allogeneic Hematopoietic Stem Cell Transplantation (HSCT). J. Clin. Oncol. 2018, 36, 7044.
[CrossRef]
83. Rambaldi, A.; Huguet, F.; Zak, P.; Cannell, P.; Tran, Q.; Franklin, J.; Topp, M.S. Blinatumomab Consolidation and Maintenance
Therapy in Adults with Relapsed/Refractory B-Precursor Acute Lymphoblastic Leukemia. Blood Adv. 2020, 4, 1518–1525.
[CrossRef] [PubMed]
84. Salhotra, A.; Yang, D.; Mokhtari, S.; Malki, M.M.A.; Ali, H.; Sandhu, K.S.; Aribi, A.; Khaled, S.; Mei, M.; Budde, E.; et al. Outcomes
of Allogeneic Hematopoietic Cell Transplantation after Salvage Therapy with Blinatumomab in Patients with Relapsed/Refractory
Acute Lymphoblastic Leukemia. Biol. Blood Marrow Transplant. 2020, 26, 1084–1090. [CrossRef] [PubMed]
85. Stock, W.; Martinelli, G.; Stelljes, M.; DeAngelo, D.J.; Gökbuget, N.; Advani, A.S.; O’Brien, S.; Liedtke, M.; Merchant, A.A.;
Cassaday, R.D.; et al. Efficacy of Inotuzumab Ozogamicin in Patients with Philadelphia Chromosome-Positive Relapsed/Refractory
Acute Lymphoblastic Leukemia. Cancer 2021, 127, 905–913. [CrossRef]
86. Jabbour, E.; Gökbuget, N.; Advani, A.; Stelljes, M.; Stock, W.; Liedtke, M.; Martinelli, G.; O’Brien, S.; Wang, T.; Laird, A.D.; et al.
Impact of Minimal Residual Disease Status in Patients with Relapsed/Refractory Acute Lymphoblastic Leukemia Treated with
Inotuzumab Ozogamicin in the Phase III INO-VATE Trial. Leuk. Res. 2020, 88, 106283. [CrossRef]
87. Marks, D.I.; Kebriaei, P.; Stelljes, M.; Gökbuget, N.; Kantarjian, H.; Advani, A.S.; Merchant, A.; Stock, W.; Cassaday, R.D.;
Wang, T.; et al. Outcomes of Allogeneic Stem Cell Transplantation after Inotuzumab Ozogamicin Treatment for Relapsed or
Refractory Acute Lymphoblastic Leukemia. Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant. 2019, 25, 1720–1729.
[CrossRef]
88. Lee III, D.W.; Stetler-Stevenson, M.; Yuan, C.M.; Shah, N.N.; Delbrook, C.; Yates, B.; Zhang, H.; Zhang, L.; Kochenderfer, J.N.;
Rosenberg, S.A.; et al. Long-Term Outcomes Following CD19 CAR T Cell Therapy for B-ALL Are Superior in Patients Receiving
a Fludarabine/Cyclophosphamide Preparative Regimen and Post-CAR Hematopoietic Stem Cell Transplantation. Blood 2016,
128, 218. [CrossRef]
89. Bassan, R.; Chiaretti, S.; Starza, I.D.; Spinelli, O.; Santoro, A.; Elia, L.; Vitale, A.; Taherinasab, A.; Piccini, M.; Ferrara, F.; et al.
Preliminary Results of the GIMEMA LAL2317 Sequential Chemotherapy-Blinatumomab Front-Line Trial for Newly Diagnosed
Adult PH-Negative B-Lineage ALL Patients. EHA Libr. 2021, 5, S114.
90. Jabbour, E.; Short, N.J.; Jain, N.; Thompson, P.A.; Kadia, T.M.; Ferrajoli, A.; Huang, X.; Yilmaz, M.; Alvarado, Y.; Patel, K.P.;
et al. Hyper-CVAD and Sequential Blinatumomab for Newly Diagnosed Philadelphia Chromosome-Negative B-Cell Acute
Lymphocytic Leukaemia: A Single-Arm, Single-Centre, Phase 2 Trial. Lancet. Haematol. 2022, 9, e878–e885. [CrossRef]
91. Litzow, M.R.; Sun, Z.; Paietta, E.; Mattison, R.J.; Lazarus, H.M.; Rowe, J.M.; Arber, D.A.; Mullighan, C.G.; Willman, C.L.;
Zhang, Y.; et al. Consolidation Therapy with Blinatumomab Improves Overall Survival in Newly Diagnosed Adult Patients with
B-Lineage Acute Lymphoblastic Leukemia in Measurable Residual Disease Negative Remission: Results from the ECOG-ACRIN
E1910 Randomized Phase 3 Nationa. Blood 2022, 140, LBA-1. [CrossRef]
92. Aldoss, I.; Forman, S.J.; Pullarkat, V. Acute Lymphoblastic Leukemia in the Older Adult. J. Oncol. Pract. 2019, 15, 67–75. [CrossRef]
93. Short, N.J.; Kantarjian, H.M.; Ravandi, F.; Huang, X.; Jain, N.; Kadia, T.M.; Khoury, J.D.; Jorgensen, J.L.; Wang, S.A.;
Alvarado, Y.; et al. Reduced-Intensity Chemotherapy with Mini-Hyper-CVD Plus Inotuzumab Ozogamicin, with or without
Blinatumomab, in Older Adults with Newly Diagnosed Philadelphia Chromosome-Negative Acute Lymphoblastic Leukemia:
Results from a Phase II Study. Blood 2020, 136, 15–17. [CrossRef]
94. Stelljes, M.; Raffel, S.; Wäsch, R.; Scholl, S.; Kondakci, M.; Rank, A.; Haenel, M.; Martin, S.; Schwab, K.; Knaden, J.; et al. First
Results of an Open Label Phase II Study to Evaluate the Efficacy and Safety of Inotuzumab Ozogamicin for Induction Therapy
Followed by a Conventional Chemotherapy Based Consolidation and Maintenance Therapy in Patients Aged 56 Years and Older
with AAcute Lymphoblastic Leukemia (INITIAL-1 trial). Blood 2020, 136, 12–13. [CrossRef]
95. Advani, A.S.; Moseley, A.; O’Dwyer, K.M.; Wood, B.L.; Fang, M.; Wieduwilt, M.J.; Aldoss, I.; Park, J.H.; Klisovic, R.B.;
Baer, M.R.; et al. SWOG 1318: A Phase II Trial of Blinatumomab Followed by POMP Maintenance in Older Patients with
Newly Diagnosed Philadelphia Chromosome-Negative B-Cell Acute Lymphoblastic Leukemia. J. Clin. Oncol. Off. J. Am. Soc.
Clin. Oncol. 2022, 40, 1574–1582. [CrossRef]
Cancers 2023, 15, 1349 23 of 23
96. Vignetti, M.; Fazi, P.; Cimino, G.; Martinelli, G.; Di Raimondo, F.; Ferrara, F.; Meloni, G.; Ambrosetti, A.; Quarta, G.;
Pagano, L.; et al. Imatinib plus Steroids Induces Complete Remissions and Prolonged Survival in Elderly Philadelphia
Chromosome-Positive Patients with Acute Lymphoblastic Leukemia without Additional Chemotherapy: Results of the Gruppo
Italiano Malattie Ematologiche Dell’Ad. Blood 2007, 109, 3676–3678. [CrossRef]
97. Foà, R.; Vitale, A.; Vignetti, M.; Meloni, G.; Guarini, A.; De Propris, M.S.; Elia, L.; Paoloni, F.; Fazi, P.; Cimino, G.; et al. Dasatinib
as First-Line Treatment for Adult Patients with Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia. Blood 2011,
118, 6521–6528. [CrossRef] [PubMed]
98. Martinelli, G.; Papayannidis, C.; Piciocchi, A.; Robustelli, V.; Soverini, S.; Terragna, C.; Marconi, G.; Lemoli, R.M.; Guolo, F.;
Fornaro, A.; et al. INCB84344-201: Ponatinib and Steroids in Frontline Therapy for Unfit Patients with Ph+ Acute Lymphoblastic
Leukemia. Blood Adv. 2022, 6, 1742–1753. [CrossRef] [PubMed]
99. Foà, R.; Bassan, R.; Vitale, A.; Elia, L.; Piciocchi, A.; Puzzolo, M.-C.; Canichella, M.; Viero, P.; Ferrara, F.; Lunghi, M.; et al.
Dasatinib–Blinatumomab for Ph-Positive Acute Lymphoblastic Leukemia in Adults. N. Engl. J. Med. 2020, 383, 1613–1623.
[CrossRef] [PubMed]
100. Jabbour, E.; Short, N.J.; Jain, N.; Huang, X.; Montalban-Bravo, G.; Banerjee, P.; Rezvani, K.; Jiang, X.; Kim, K.H.;
Kanagal-Shamanna, R.; et al. Ponatinib and Blinatumomab for Philadelphia Chromosome-Positive Acute Lymphoblastic
Leukaemia: A US, Single-Centre, Single-Arm, Phase 2 Trial. Lancet Haematol. 2023, 10, e24–e34. [CrossRef]
101. Webster, J.; Luskin, M.R.; Prince, G.T.; DeZern, A.E.; DeAngelo, D.J.; Levis, M.J.; Blackford, A.; Sharon, E.; Streicher, H.;
Luznik, L.; et al. Blinatumomab in Combination with Immune Checkpoint Inhibitors of PD-1 and CTLA-4 in Adult Patients with
Relapsed/Refractory (R/R) CD19 Positive B-Cell Acute Lymphoblastic Leukemia (ALL): Preliminary Results of a Phase I Study.
Blood 2018, 132, 557. [CrossRef]
102. Schwartz, M.; Damon, L.E.; Jeyakumar, D.; Costello, C.L.; Tzachanis, D.; Schiller, G.J.; Reiner, J.; Wieduwilt, M.J. Blinatumomab in
Combination with Pembrolizumab Is Safe for Adults with Relapsed or Refractory B-Lineage Acute Lymphoblastic Leukemia:
University of California Hematologic Malignancies Consortium Study 1504. Blood 2019, 134, 3880. [CrossRef]
103. Ghorashian, S.; Kramer, A.M.; Onuoha, S.; Wright, G.; Bartram, J.; Richardson, R.; Albon, S.J.; Casanovas-Company, J.; Castro, F.;
Popova, B.; et al. Enhanced CAR T Cell Expansion and Prolonged Persistence in Pediatric Patients with ALL Treated with a
Low-Affinity CD19 CAR. Nat. Med. 2019, 25, 1408–1414. [CrossRef] [PubMed]
104. Amrolia, P.J.; Wynn, R.; Hough, R.E.; Vora, A.; Bonney, D.; Veys, P.; Chiesa, R.; Rao, K.; Clark, L.; Al-Hajj, M.; et al. Phase I Study
of AUTO3, a Bicistronic Chimeric Antigen Receptor (CAR) T-Cell Therapy Targeting CD19 and CD22, in Pediatric Patients with
Relapsed/Refractory B-Cell Acute Lymphoblastic Leukemia (r/r B-ALL): Amelia Study. Blood 2019, 134, 2620. [CrossRef]
105. Kantarjian, H.; Jabbour, E. Incorporating Immunotherapy into the Treatment Strategies of B-Cell Adult Acute Lymphoblastic
Leukemia: The Role of Blinatumomab and Inotuzumab Ozogamicin. Am. Soc. Clin. Oncol. Educ. Book Am. Soc. Clin. Oncol. Annu.
Meet. 2018, 38, 574–578. [CrossRef] [PubMed]
106. Gaballa, M.R.; Banerjee, P.; Milton, D.R.; Jiang, X.; Ganesh, C.; Khazal, S.; Nandivada, V.; Islam, S.; Kaplan, M.; Daher, M.; et al.
Blinatumomab Maintenance after Allogeneic Hematopoietic Cell Transplantation for B-Lineage Acute Lymphoblastic Leukemia.
Blood 2022, 139, 1908–1919. [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.