You are on page 1of 13

HEMATOLOGIC MALIGNANCIES

Recent Advances in Managing Acute


Lymphoblastic Leukemia
Daniel J. DeAngelo, MD, PhD1; Elias Jabbour, MD2; and Anjali Advani, MD3
overview

Acute lymphoblastic leukemia (ALL) is characterized by chromosomal translocations and somatic mutations
that lead to leukemogenesis. The incorporation of pediatric-type regimens has improved survival in young
adults, and the incorporation of tyrosine kinase inhibitors for patients with Philadelphia chromosome–positive
disease has led to further improvements in outcomes. However, older patients often have poor-risk biology and
reduced tolerance to chemotherapy, leading to lower remission rates and overall survival. Regardless of age,
patients with relapsed or refractory ALL have extremely poor outcomes. The advent of next-generation se-
quencing has facilitated the revolution in understanding the genetics of ALL. New genetic risk stratification
together with the ability to measure minimal residual disease, leukemic blasts left behind after cytotoxic
chemotherapy, has led to better tools to guide postremission approaches—that is, consolidation chemo-
therapy or allogeneic stem cell transplantation. In this article, we discuss the evolving and complex genetic
landscape of ALL and the emerging therapeutic options for patients with relapsed/refractory ALL and older
patients with ALL.

INTRODUCTION Regardless of age, relapsed disease is an insur-


ALL is a rare disease, with 5,930 new cases diagnosed mountable problem for many patients. The higher rate
in the United States in 2019 (0.3% of all cancers).1 ALL of relapse among adults with ALL is probably attrib-
is increasingly recognized as a genetically heteroge- utable to higher-risk disease. For example, more adult
neous disease, and a flurry of new genetic subtypes has patients have adverse cytogenetics and molecu-
led to a refinement in risk stratification. Although most lar features (i.e., the Philadelphia chromosome or
cases of ALL are diagnosed in the pediatric population, Philadelphia-like signature) and persistent MRD. Sal-
the incidence follows a bimodal pattern, with the first vage chemotherapy regimens have shown only modest
peak occurring in children younger than age 5 and activity for patients with relapsed or refractory ALL.14,15
a second peak occurring at approximately age 50.2 Monoclonal antibodies, novel immunotherapies, and
Measurement of minimal residual disease (MRD) has kinase inhibitors for specific genetic subtypes are
led to prompt risk-adapted interventions and improved entering the clinic in the upfront and relapsed/
outcomes.3,4 Tremendous progress has been made in refractory (R-R) setting, with promising results.
the treatment of children with ALL, with complete re-
mission (CR) rates of 95% and estimated 5-year event- HOW DO I PROFILE ALL WITH CURRENT
free survival (EFS) rates of 80% to 85%.5-7 Unfortunately, MOLECULAR DATA?
the results for adult patients have not kept up with Risk stratification by cytogenetics and molecular ge-
those of their pediatric counterparts. Despite CR rates netics reflects the prognostic heterogeneity of ALL that
of approximately 85% with adult regimens, the 3-year determines which patients are at a high risk of relapse
Author affiliations EFS and overall survival (OS) rates remain below and should be considered for more intensive treat-
and support
45%.8,9 Pediatric-inspired regimens that rely heavily ment strategies, including allogeneic stem cell trans-
information (if
applicable) appear
on intensification of corticosteroids, pegylated aspar- plantation (ASCT). Risk assessment at diagnosis based
at the end of this aginase, vinca alkaloids, and antimetabolites are on immunophenotypic and genetic factors will allow
article. currently being used for young adult patients, leading stratification of patients into standard-risk and high-risk
Accepted on March to improvements in the EFS and OS rates when categories. In addition, patient characteristics, in-
2, 2020 and compared with historical controls.10-13 However, these cluding advanced age, poor performance status, and
published at
agents are poorly tolerated by older patients, resulting hyperleukocytosis, are all recognized adverse risk
ascopubs.org on May
18, 2020: DOI https://
in poor survival rates. Therefore, newer and lower- factors predicting poor outcomes of chemotherapy. In
doi.org/10.1200/ intensity therapies incorporating novel immunother- both pediatric and adult studies, persistence of MRD,
EDBK_280175 apies and now being tested for older patients with ALL. defined as more than 10 4 blasts, has proven to be

330 2020 ASCO EDUCATIONAL BOOK | asco.org/edbook

Downloaded from ascopubs.org by 2.57.77.86 on May 19, 2020 from 002.057.077.086


Copyright © 2020 American Society of Clinical Oncology. All rights reserved.
Recent Advances in Managing Acute Lymphoblastic Leukemia

somatic mutations typical of myeloid malignancies.27 In


a retrospective analysis, 17% of adult patients with T-ALL or
PRACTICAL APPLICATIONS
lymphoblastic lymphoma were found to have the charac-
• Cytogenetics and molecular profiling are nec- teristic ETP-ALL immunophenotype. These patients, when
essary for appropriate risk stratification in pa-
treated with hyper-CVAD (i.e., hyperfractionated cyclo-
tients with ALL.
phosphamide, vincristine, doxorubicin, and dexametha-
• Assessment of the Philadelphia-like signature is sone) had significantly lower remission rates (73% vs. 91%;
essential for decisions regarding postremission p = .03) and a shorter median OS (20 months vs. not
therapy and treatment at relapse.
reached; p = .008).28 In contrast, in a cohort of pediatric and
• The assessment of minimal residual disease adolescent patients treated with a pediatric regimen (Chil-
remains the most important assessment for risk dren’s Oncology Group AALL0434), ETP-ALL was not as-
stratification in ALL. sociated with adverse outcomes, but postremission therapy
• Novel immunotherapy agents are more effective was based on MRD status.29
than conventional therapy for patients with re-
lapsed or refractory disease. Cytogenetic Classification
• The treatment of older patients with ALL re- The most common chromosomal abnormalities observed in
mains a difficult endeavor with high therapy-
children with B-cell ALL are hyperdiploidy (. 50 chromo-
related toxicity and poor long-term survival, and
somes) and the ETV6-RUNX1 (t12;21) subtypes, both of
therefore novel approaches are needed.
which are associated with favorable outcomes (Table 1).30
Adolescent and young adult patients with ALL have a much
a major independent risk factor for relapse.16 Thus, MRD lower incidence of hyperdiploidy and ETV6-RUNX131 and
assessment by either molecular- or flow-based approaches a higher incidence of unfavorable cytogenetics, including
has become standard clinical practice not only for pediatric Philadelphia-positive ALL, hypodiploidy, and complex kar-
but also for adult patients.17 yotype. The most important cytogenetic abnormality is the
Philadelphia chromosome. It is seen in only 3% of pediatric
Immunophenotypic Classification patients but approximately 25% of adults, and it increases
Determining the cell of origin (T-cell vs. B-cell ALL) and with age, representing approximately half of the cases in
specific immunophenotypes (e.g., Pro-B and Pre-B ALL) patients older than age 60.32 Although the outcome of
were traditionally of prognostic importance in ALL18-20; patients with Philadelphia chromosome positive–ALL has
however, more recent reports dispute their prognostic improved with the addition of a tyrosine kinase inhibitor
value.21 It remains essential to separate precursor B-cell (TKI), it remains unclear whether young adults need an
ALL from mature B-cell ALL (Burkitt) because these entities intensive chemotherapy (IC) backbone and whether ASCT
are typically treated with different chemotherapy strategies. is still necessary for patients who achieve molecular re-
Mature B-cell tumors typically express surface immu- mission. Single-agent imatinib or dasatinib plus cortico-
noglobulin and are negative for CD34 and terminal steroid therapy, pioneered by the Gruppo Italiano Malattie
deoxytransferase markers of immaturity. In B-cell ALL, Ematologiche dell’Adulto (GIMEMA),33,34 induced CR in
the expression of CD20 (. 20%) has consistently been almost all patients without risk of induction death. With TKI
shown to be associated with disease resistance and poorer plus chemotherapy combinations, CR rate exceeded 95%,
outcomes,22,23 which can be ameliorated with the adminis- but death occurred in 2% to 7% of the cases. In a ran-
tration of anti-CD20 immunotherapies, such as rituximab.24 domized trial from the Group for Research on Adult Acute
T-cell ALL accounts for approximately 15% of ALL, and with Lymphoblastic Leukemia,35 a combination of de-escalated
pediatric-based regimens adopting an MRD risk-stratified chemotherapy and TKI resulted in less induction toxicity
approach, the outcomes of T-cell ALL may be superior to and noninferior CR and survival results compared with
those of B-cell ALL. Subgroup analysis from the large standard chemotherapy plus TKI. In an MD Anderson
Medical Research Council UKALLXII/Eastern Cooperative Cancer Center study, ponatinib combined with hyper-CVAD
Oncology Group (ECOG) 2993 trial demonstrated that, in led to an excellent 83% 2-year OS, even without ASCT.36
T-cell ALL, CD1a positivity with the absence of CD13 ex- KMT2A (also known as the mixed lineage leukemia
pression was associated with improved survival. 25 Early gene [MLL] on 11q23)–rearranged ALL carries a poor
T-cell precursor (ETP) ALL is a recently recognized prognosis.30,37 The incidence of KMT2A ranges from 60% to
immunophenotypic high-risk subgroup of T-cell ALL. ETP 80% in infants with ALL. In children and adults, the in-
ALL has a characteristic immunophenotype characterized cidence of KMT2A gene rearrangements ranges from 4.5%
by CD1aneg, CD8neg, and CD5weak with aberrant expression of to 5.7%. One of the most common 11q23 abnormalities,
myeloid markers.26 Interestingly, ETP-ALL often harbors t(4;11), occurs in 2% of children and adults with ALL.

2020 ASCO EDUCATIONAL BOOK | asco.org/edbook 331

Downloaded from ascopubs.org by 2.57.77.86 on May 19, 2020 from 002.057.077.086


Copyright © 2020 American Society of Clinical Oncology. All rights reserved.
DeAngelo, Jabbour, and Advani

TABLE 1. Common Chromosomal and Molecular Abnormalities in B-Cell ALL


Cytogenetics Gene Incidence in Adults Incidence in Children
Hyperdiploidy (. 50 chromosomes) — 7% 25%–30%
t(12;21)(p13;q22) ETV6-RUNX1 (TEL-AML1) 2% 22%–25%
t(9;22)(q34;q11): Philadelphia chromosome BCR-ABL1 25% 2%–4%
t(4:11)(q21;q23) and other KNMT2A translocations KNMT2A 8%–10% 2%–3% (60%–80% in infants)
Low hypodiploid near triploid TP53 in low hypodiploid 8%–10% 2%–3%
t(1;19)(q23;p13) TCF3-PBX1 3% 4%
t(11;14)(q11), e.g., (p13;q11), (p15;q11) TCRα and TCRδ 20%–25% 10%–20%
BCR-ABL1-like, Philadelphia-like Various 10%–30% 15%
Ikaros IKZF1 25%–35% 12%–17%

KMT2A rearrangements seldom express CD10, and patients with Philadelphia-like ALL have a poorer outcome
therefore the blasts have a common immunophenotype of compared with other B-cell ALLs, and it remains unclear
CD19+, CD10 , CD15+, or CD65+. Interestingly, they are whether they should receive ASCT up front or based on
associated with high levels of H3K79 methylation catalyzed MRD persistence.45 The Children’s Oncology Group is
by the DOTL1 enzyme, suggesting a novel therapeutic testing ruxolitinib in patients with CRLF2-rearranged and
target.38,39 In addition, multiple studies have shown high JAK-STAT dysregulated disease (NCT02723994).
levels of FLT3 expression in patients with KMT2A rear- The most common mutation in classic T-cell ALL is
rangements; therefore, inhibitors of FLT3 (a tyrosine kinase) NOTCH1, whereas DNMT3A and other myeloid-specific
may also prove beneficial.40 In some patients, KMT2A gene mutations are more commonly seen in ETP-ALL.27 NOTCH1
rearrangements are not detected by conventional cytoge- can be targeted by the gamma-secretase inhibitors, which
netics, and commercially available dual-color KMT2A have been tested in patients with advanced late-stage
probes can detect translocations in metaphase chromo- disease, with some durable responses but at the expense of
somes by fluorescence in situ hybridization techniques. gut toxicity.48
TCF3-PBX1, associated with t(1;19) translocation, is char-
Minimal Residual Disease
acterized by favorable outcomes with intensive treat-
ment. Hypodiploid ALL is a poor prognostic subtype, which Patients with detectable residual blasts after chemotherapy
includes near-haploid (24–31 chromosomes), low-hypodiploid (MRD positive, typically defined as more than 10 4 blasts)
(32–39 chromosomes), and high-hypodiploid (40–43 chro- are seldom cured with chemotherapy alone. In pro-
mosomes) groups. RAS and PI3K pathways are frequently spective trials,17,49,50 the OS was between 60% and 80%
mutated in near-haploid ALL, whereas TP53 and IKZF are with chemotherapy alone in patients with MRD-negative
often mutated in low-hypodiploid ALL.41 Therefore, germline status, even in high-risk subsets and Ph-positive ALL.
mutational screening of TP53 should always be performed in Conversely, patients with MRD-positive status benefit par-
these cases. tially from ASCT but with OS rates of 50% or less in intention-
to-treat analyses because of the cumulative effects of
Molecular Classification pretransplantation and post-transplantation relapse and
The Philadelphia-like subgroup, initially identified by means transplant-related deaths.4,51,52
of gene expression profiling, accounts for approximately Monitoring MRD with real-time quantitative polymerase
20% of young adult and adult B-cell ALL cases (Table 1). chain reaction, flow cytometry, or high-throughput–based
These cases are characterized by a transcriptional profile platforms should be considered a standard practice for the
similar to that of Philadelphia-positive ALL but lack the t(9;22)/ treatment of patients with ALL because it is the best pre-
BCR-ABL1 rearrangement.42-45 Instead, the underlying ge- dictor of OS. Similar robust prognostic data are seen in both
nomic lesions are heterogeneous, making its recognition B-cell and T-cell ALL. In the analysis of standard-risk pa-
difficult and uneven between trials. Rearrangements in tients enrolled on the German Multicenter Study Group for
CRLF2 are seen in approximately 50% of cases; rear- Acute Lymphoblastic Leukemia study, early MRD clearance
rangements in the ABL class genes (ABL1, ABL2, CSF1R, before day 24 was associated with a favorable outcome
PDGFRA, PDGFRB), in roughly 10%; and JAK/STAT genes (relapse rate of 0% at 3 years), whereas MRD positivity
(JAK1-3, IL7R, and CRLF2 mutations), less often (, 10%). lasting beyond week 16 predicted a relapse rate of 94%.53
Deletions in IKZF1 occur in up to 80% of cases and are Therefore, patients who achieve CR but fail to achieve
associated with a particularly poor prognosis.46,47 In general, a negative MRD status remain at high risk of early relapse

332 2020 ASCO EDUCATIONAL BOOK | asco.org/edbook

Downloaded from ascopubs.org by 2.57.77.86 on May 19, 2020 from 002.057.077.086


Copyright © 2020 American Society of Clinical Oncology. All rights reserved.
Recent Advances in Managing Acute Lymphoblastic Leukemia

and should seek alternative therapy.4 Similarly, MRD status binding region.62,63 In the confirmatory phase II study of 189
before SCT is also a powerful indicator for post-transplantation patients with R-R Philadelphia-negative ALL, blinatumomab
outcomes and disease resistance.54 was associated with a CR-plus–CR with partial hematologic
MRD is such a powerful predictor for relapse, so it is cur- recovery rate of 43%. The median response duration was 9
rently used to guide postinduction decisions. Bassan et al55 months, and the median OS was 6 months.64
allocated patients to maintenance or SCT based on MRD After these results, a phase III trial (TOWER study) was
status at the end of intensive therapy (approximately conducted: more than 400 patients with R-R Philadelphia-
5 months from attaining CR) regardless of their cytogenetic negative ALL were randomly assigned (2:1) to blinatumo-
and clinical risk score at diagnosis, resulting in an im- mab (n = 271) or standard of care (n = 134; Table 2).59 The
pressive 75% 5-year OS rate for nontransplanted patients response rates were 44% and 25%, respectively (p , .001).
attaining MRD-negative status. This approach is being used Molecular remission rates among responders, defined as
by other large cooperative groups with similar success.56 fewer than 10 4 blasts in the first 12 weeks, were 76% and
Taken together, these data suggest that MRD may indeed 48%, respectively. Blinatumomab prolonged survival, the
be the most important predictor of OS for patients with ALL. primary study endpoint; the median OS was 7.7 months
Future Directions (95% CI, 5.6–9.6) versus 4.0 months (95% CI, 2.9–5.3; p =
.01; hazard ratio, 0.71). A total of 24% of the patients in
We are entering an intensive phase of clinical investigations each group underwent ASCT.65
using next-generation sequencing data for the exploration of
individualized or subset-specific treatment algorithms. A recent phase III study demonstrated the superiority of
Therefore, it will be crucial to design prospective clinical blinatumomab for 208 children and adolescent young
studies with modular data to evaluate optimal strategies adults in salvage 1 and randomly assigned (1:1) to bli-
for specific patient subtypes, or “personalized” medicine, natumomab or standard of care.66 Blinatumomab induced
testing immunotherapy and combinations of molecularly a higher rate of measurable residual disease negativity (79%
targeted drugs or drug combinations. vs. 21%; p , .001) and a higher 2-year OS (79% vs. 59%;
p = .005).
TREATING RELAPSED-REFRACTORY ALL: TOO MANY
OPTIONS, WHICH ONE TO PICK? Blinatumomab was evaluated in the phase II ALCANTARA
trial with 45 patients who had R-R Philadelphia-positive
Therapies targeted toward specific transcripts (e.g., BCR- ALL.67 Thirty-six percent achieved a response. The median
ABL1 tyrosine kinase oncoprotein by TKIs) and specific relapse-free survival and OS were 6.7 months and 7.1
leukemic cell surface antigens (e.g., CD20, CD22, and months, respectively; 44% of patients underwent ASCT.
CD19 monoclonal antibodies) are major breakthroughs in Furthermore, blinatumomab was given with TKIs to 20 pa-
the management of ALL.57 Historically, relapsed or re- tients with R-R Philadelphia-positive ALL or chronic myeloid
fractory (R-R) ALL was associated with a dismal prognosis, leukemia in the lymphoid blast phase. The response rate was
with a cure rate of less than 10%.14,58 The CR rates with 65%. The median survival was 14 months.68
standard chemotherapy regimens are 30% to 40% in first
relapse and 20% to 25% in second relapse. Accordingly, Gökbuget et al69 assessed blinatumomab in 113 patients
only 10% to 30% adult patients with relapsed ALL proceed with ALL in MRD-positive CR. Approximately 78% of patients
to ASCT, which is the only curative option in this setting.14 achieved MRD negativity after one cycle and 80 after two
Recent developments of monoclonal antibodies, bispecific cycles. With a median follow-up of 29 months, the median OS
antibody constructs, and CAR T-cell therapies have revo- was 36.5 months and the relapse-free survival was 18.9
lutionized the treatment of ALL, resulting in U.S. Food and months. The median OS with achievement of MRD negativity
Drug Administration (FDA) approvals of blinatumomab in was 38.9 months compared with 12.5 months for patients with
2014 and inotuzumab and tisagenlecleucel in 2017 as ALL persistent MRD. These findings were confirmed when com-
salvage strategies.59-61 Their use in combination with other pared with historical data via propensity score matching.70
treatment modalities in the salvage and frontline settings are Blinatumomab is being evaluated in the frontline setting
under investigation. in combination with chemoimmunotherapy in Philadelphia-
negative ALL and with TKIs in Philadelphia-positive ALL.
Anti-CD19 Bispecific T-Cell Engager:
Blinatumomab Anti-CD22 Antibody-Drug Conjugate:
Blinatumomab is an anti-CD19–directed CD3 bispecific T-cell Inotuzumab Ozogamicin
engager constructed with BiTE antibody technology.62-64 It Inotuzumab ozogamicin is a novel anti-CD22 monoclonal
consists of a recombinant monoclonal antibody com- antibody conjugated to the toxin calicheamicin.71 In a single-
posed of an anti-CD19 fragment antigen-binding region institution, phase II study of patients with R-R ALL, inotu-
joined by a short link to an anti-CD3 fragment antigen- zumab was administered at a starting intavenous dosage of

2020 ASCO EDUCATIONAL BOOK | asco.org/edbook 333

Downloaded from ascopubs.org by 2.57.77.86 on May 19, 2020 from 002.057.077.086


Copyright © 2020 American Society of Clinical Oncology. All rights reserved.
DeAngelo, Jabbour, and Advani

TABLE 2. Relapsed and Refractory Trials in ALL


Median Overall Survival Minimal Residual Disease
Clinical Trial Treatment Setting Patients Response (months) Negativitya
Blinatumomab
TOWER4 R-R Ph ALL 271 ORR: 44% 7.7 76%
12
ALCANTARA R-R Ph+ ALL 45 ORR: 36% 7.1 88%b
Inotuzumab Ozogamicin
INO-VATE5 R-R Ph– and Ph+ 164 ORR: 74% 7.7 78%
ALL
Inotuzumab plus mini-hyper-CVD 6 R-R Ph– ALL 89 ORR: 79% S1: 25 82%
blinatumomab23
S2: 6
S3: 7
CAR T
ELIANA82 R-R ALL 79 ORR: 81% NR 100%

Abbreviations: ALL, acute lymphoblastic leukemia; mini-hyper-CVD, mini-hyperfractionated cyclophosphamide, vincristine, and dexamethasone; NR, not
reached; Ph, Philadelphia chromosome; ORR, overall response rate; R-R, relapsed or refractory; S1, salvage 1; S2, salvage 2; S3, salvage 3.
a
Measured by flow cytometry.
b
Within two cycles.

1.3 to 1.8 mg/m2 every 3 to 4 weeks. Forty-nine patients were 65%, respectively. The 12-month survival rate was 40%.
treated.72 The objective response rate was 57%, and the The VOD rate was 10.4%; it was 26% after ASCT.
median survival was 5.1 months. Nearly half of the patients Inotuzumab was also evaluated with a dose-reduced mini-
treated with inotuzumab proceeded to ASCT. To minimize hyper-CVD regimen (i.e., mini-hyperfractionated cyclo-
toxicities and based on pharmacokinetic and pharmaco- phosphamide, vincristine, and dexamethasone) with or
dynamic data, inotuzumab was administered intravenously without blinatumomab in 89 patients with R-R ALL.77 The
on a weekly basis at 0.8 mg/m2 on day 1, followed by 0.5 mg/ response rate was 79%, with 82% of responders achieving
m2 on days 8 and 15, every 3 to 4 weeks in 40 patients. The MRD negativity. The 2-year progression-free survival and OS
study yielded a similar response rate to inotuzumab given rates were 52% and 39%, respectively. Among patients
every 3 to 4 weeks (59% vs. 57%), with a median survival of treated in salvage 1, the 2-year survival rate was 51%. The
9.5 months.73 Weekly administration of inotuzumab resulted VOD rate was 6%.
in fewer adverse events, including lower rates of veno-
occlusive disease (VOD). In a separate multicenter phase Better outcomes were obtained in patients who received
II trial in heavily pretreated patients with R-R ALL, inotu- inotuzumab- or blinatumomab-based therapies in first
zumab therapy resulted in a remission rate of 66%, with 78% salvage.65,78-81 This was particularly notable in patients who
of patients who achieved CR becoming MRD negative. The achieved a negative MRD status and received subsequent
median survival was 7.4 months.74 ASCT. ASCT performed in subsequent salvages did not
confer a survival advantage.80
These results led to a randomized trial comparing inotu-
zumab with physician’s choice of chemotherapy in R-R CAR T-Cell Therapies
ALL.60,74 The response rate was 88% (CR, 81%) with ino- CAR T Cells are genetically modified autologous T lym-
tuzumab and 32% (CR, 29%) with standard of care (p , phocytes engineered to express binding sites of specific
.0001). Among responders, the MRD negativity rates were antibodies, such as a receptor against CD19. These T cells,
78% and 28% (p , .0001), respectively. The median harnessed from the patients’ own immune systems, target
survival was 7.7 versus 6.7 months (p = .02; hazard ratio, the malignant cells.
0.77); The 2-year survival rate was 23% versus 10%.75 In a phase II multicenter study, 79 children and young
Serious toxicities included VOD after ASCT, mainly in pa- adults with relapsed or refractory CD19+ B-ALL received
tients who received double alkylators in pretransplantation a single infusion of tisagenlecleucel, CD19 CAR T-cells.61,82
conditioning. Age was also a risk factor for VOD. Among evaluable patients, the overall response rate was
Inotuzumab was assessed in 48 children, adolescents, and 81%, with all patients whose disease responded achieving
young adults (median age, 9; range, 1–21 years).76 The negative MRD. The 24-month relapse-free survival and OS
objective response and MRD negativity rates were 62% and rates were 62% and 66%, respectively. The CAR T cells

334 2020 ASCO EDUCATIONAL BOOK | asco.org/edbook

Downloaded from ascopubs.org by 2.57.77.86 on May 19, 2020 from 002.057.077.086


Copyright © 2020 American Society of Clinical Oncology. All rights reserved.
Recent Advances in Managing Acute Lymphoblastic Leukemia

persisted for a median of 168 days (range, 20–617 days), agents have demonstrated single-agent and synergistic cell
suggesting that immunosurveillance with these products killing when combined with chemotherapy because of the
may be long lasting. Grade 3 and 4 cytokine release syn- upregulation of Bcl-2 in these cell lines.89,90 In a phase I trial,
drome (CRS) and neurotoxicities were encountered in 49% 36 patients with R-R ALL (median age, 27; range, 6–72
and 40%, respectively, with 48% needing an intensive care years) were treated with venetoclax and navitoclax in
unit stay. This led to the approval of tisagenlecleucel for R-R combination with chemotherapy. The objective response
ALL for patients up to age 25 after failure of two previous rate was 50%; the 6-month CR duration rate was 43%.92
therapies. Preliminary results of the combination of venetoclax with
low-intensity chemotherapy in newly diagnosed older pa-
For 53 adults with heavily pretreated B-ALL who received
tients unfit for IC are promising, with objective response and
the CAR T-cell infusion, the CR rate was 83%, including 32
MRD negativity rates of 91% and 100%, respectively.93
(67%) of 48 evaluable patients achieving negative MRD.83
Better outcomes were observed for patients with low disease Future Directions
burden ( 5% bone marrow blasts) at the time of CAR T-cell
Given the encouraging results achieved with monoclonal
infusion, with a median EFS of 10.6 months compared with
antibodies, bispecific antibody constructs, and CAR T cells,
5.3 months and a median OS of 20.1 months compared
the therapeutic tools necessary to improve outcomes of
with 12.4 months for patients with a high disease burden.
patients with adult ALL may now be available. These
Several strategies to optimize CAR T-cell activities and treatment modalities are not competitive but rather com-
minimize their toxicities have been explored. The use of plementary, and they could be administered sequentially to
CD19 CAR T cells with a lower-affinity CD19 binder with produce the deepest remissions possible. Their rational
a fast off-rate would lead to physiologic T-cell activation, combination in the frontline setting is ongoing and may
reduced toxicity, improved engraftment, and potential long- reduce the need for long-term IC and obviate ASCT for many
term persistence to deliver sustained responses.84 In a pilot patients.
trial of 16 adults treated, the objective response and MRD
negativity rates were 87% each. The 6-month OS rate was THE OLDER PATIENT WITH ALL
66%. No grade 3 cases of CRS were reported; three patients The definition of “older” or “elderly” adults with ALL has
experienced reversible grade 3 neurotoxicity.85 typically included patients age 60 and older. Historically,
their prognosis has been dismal, with a 5-year OS of ap-
To circumvent CD19 escape as a cause of relapse after
proximately 20%.94,95 This prognosis has been attributed to
CD19 CAR T-cell therapy, CD22-targeted CAR T-cell ther-
adverse biology and the inability of older patients to tolerate
apy has been developed.86 Of the 15 children and adults
IC. However, the advent of new immunotherapies and TKIs,
with R-R B-ALL, most of whom were previously treated with
incorporation of MRD to guide therapy, and the increasing
CD-19–directed immunotherapy, 11 (73%) achieved CR
safety of ASCT for older patients are leading to improved
after treatment with at least 1  106/kg body weight of CAR
outcomes. However, these new approaches make man-
T cells. Encouraging results from a phase I trial of dual
agement of these cases more complicated. In this section,
CD19/CD22 bispecific CAR T-cells in 21 children and adults
we focus on disease biology, standard treatment, and new
with R-R ALL reported grade 3 to 4 CRS and neurotoxicity in
approaches to therapy in the older patient with ALL.
only two patients (10%), with CR and MRD negativity rates
of 86% and 81%, respectively.87 Biologic Concerns for Older Patients With ALL
Current therapies use autologous lymphocytes, which can The incidence of high-risk genetic abnormalities among
be scarce and difficult to expand. New platforms provide an older adults with ALL is high. The Philadelphia chromosome
off-the-shelf approach, with cells derived from healthy is present in approximately 50% of these patients.94,96
volunteer donors. In the phase I dose escalation trial in Before the advent of TKIs, the Philadelphia chromo-
children and adults with R-R ALL, the objective response some was associated with a poor prognosis; however, TKIs
and MRD negativity rates were 82% and 71%, re- have dramatically improved outcomes.97 More recently,
spectively.88 Treatment was tolerated, with only moderate the Philadelphia-like signature has been identified in
CRS. Off-the-shelf products targeting CD22 and allogeneic approximately 24% of older adults and has been asso-
cord blood–derived natural killer cells are being developed. ciated with a poor outcome.43 The incidence of specific
fusions varies with age, with older adults expressing high
BH3 Mimetics levels of CRLF2.43 Low hypodiploidy or near triploidy,
Preclinical studies have demonstrated activity of the BH3 complex cytogenetics, IKZF1 mutations, chromosome 17
mimetics venetoclax and navitoclax in B-cell and T-cell ALL abnormalities, and KMT2A rearrangements are more
cell lines.89-91 Venetoclax has demonstrated particularly common, and high hyperdiploidy is less common, in older
strong activity in MLL-rearranged B-cell ALL, and both adults with ALL.98-103 Finally, older patients have a higher

2020 ASCO EDUCATIONAL BOOK | asco.org/edbook 335

Downloaded from ascopubs.org by 2.57.77.86 on May 19, 2020 from 002.057.077.086


Copyright © 2020 American Society of Clinical Oncology. All rights reserved.
DeAngelo, Jabbour, and Advani

incidence of the B-cell (89%) compared with T-cell (66%) removing asparaginase and cyclophosphamide.94,110 Al-
phenotype.98,104 though targeting CD20 with rituximab in combination with
chemotherapy has demonstrated benefit in patients younger
Outcomes of Standard Treatment for Patients With than age 60 with B-ALL, incorporating this agent for patients
Philadelphia Chromosome–Negative ALL who are at least age 60 has not demonstrated a benefit.94,111
Standard ALL treatment includes intensive induction and This finding may be related to more deaths in CR. Ribera
postremission therapy. This can be challenging for older et al112 evaluated a pediatric-inspired IC in a younger group of
adults because of their comorbidities: diabetes, cardiac older adults (age 55–65) to see whether results were more
dysfunction, pulmonary abnormalities, neuropathy, poly- favorable in this subgroup. Despite an improvement in out-
pharmacy, and decline in performance status.94,98 Although come, the 2-year EFS was only 37%,112 emphasizing the
poor outcomes have been reported in clinical trials, out- need for novel strategies.
comes are almost certainly worse when evaluating “all
comers” because many patients do not receive treatment. Patients With Philadelphia Chromosome–Positive ALL
In a Surveillance, Epidemiology, and End Results data- TKI treatment has improved the outcomes for Philadelphia
base of patients age 60 or older with ALL, the median OS chromosome–positive ALL substantially. First-generation
was 4 months, and the 3-year OS was 12.8%.98,105 In the TKIs, such as imatinib, have demonstrated higher rates
largest study conducted with IC for older adults with of remission than chemotherapy alone and low rates of
Philadelphia-negative ALL, the German Multicenter toxicity.98,113 However, remission durations have typically
Study Group for Acute Lymphoblastic Leukemia trial,98,106 been short (median duration, 8 months with imatinib).34
268 patients older than age 55 were treated with a modified Later-generation TKIs, such as dasatinib, have demon-
intensive Berlin-Frankfurt-Munster (i.e., BFM) regimen strated improved results.33 However, despite all patients
followed by postremission therapy and maintenance. Rit- achieving a hematologic remission, only 20% achieved
uximab was administered to patients with CD20-positive a major molecular remission, and the 20-month OS and
disease. The 5-year OS was only 23% despite a relatively disease-free survival rates were 69.2% and 51.1%, re-
young median age (67 years) and 62% of patients having an spectively.33 Twelve of the 17 patients who experienced
ECOG performance status of 0 to 1. An amendment was relapse had the resistant T315I Bcr-Abl mutation,33 the
incorporated into the trial, adding triple intrathecal che- dominant mechanism of relapse among patients treated
motherapy, adding asparaginase during postremission with earlier-generation TKIs.33,98,114 Ponatinib is the only
therapy, and increasing the dosages of methotrexate and active TKI in this setting. The GIMEMA group evaluated
cytarabine. The 2-year OS increased from 33% to 52%; ponatinib in combination with steroids.98,115 The rates of CR,
however, long-term outcomes remained disappointing.98,106 complete molecular remission, and 1-year OS were 95%,
With most IC, the rates of CR have decreased with ad- 46%, and 87.5%, respectively. Thirteen serious adverse
vancing age and the rates of early death have increased events related to ponatinib occurred (in 44 patients).94,115
(Table 3).106 Increased age has been associated with Therefore, caution is necessary, given the cardiovascular
a higher risk of infections during induction (81% vs. 70%) risk profile of ponatinib, and the drug dose should be re-
and more dose reductions (46% vs. 28%).107 In patients age duced once complete molecular remission is achieved.
60 or older treated with hyper-CVAD, the death rate in CR These results are encouraging, however, the median follow-
has been approximately 34%.94,108 Because of the in- up is short (11.4 months). Longer follow-up is needed.
creased rates of early deaths, most IC regimens have used
For fit older adults, TKI therapy can be combined with IC.
a standard backbone with dosage reductions to avoid tox-
Imatinib has been combined effectively with various che-
icities. The Cancer and Leukemia Group B reduced the
motherapy regimens: UKALLXII/ECOG 2993, the North
number of days of steroids to 7 days (from 21 days) during
Italian Leukemia Group multiagent chemotherapy back-
induction for older patients.109 In a PETHEMA trial, re-
bone, GRAAPH-2005 with reduced-intensity induction, and
searchers increased the 2-year OS from 39% to 52% by
hyper-CVAD.35,116-119 Although the CR rates have been more
than 90%, higher than those of chemotherapy alone, the
TABLE 3. Rates of Complete Remission and Early Death by Age Group rates of EFS and OS have varied (EFS, 33%–42%; OS,
in the German Multicenter Study Group for Acute Lymphoblastic 38%–48%).35,116-119 This variation may be related to the
Leukemia Study14 regimen, median patient age, and percentage of patients
Age Range (years) Complete Remission Rate Early Death Rate ultimately undergoing ASCT. Notably, the median age of
55–65 84% 7% patients in most of these trials was in the mid-40s, and OS
66–75 74% 14%
was significantly shorter in patients age 60 or older,
compared with those younger than age 60, stressing the
. 75 52% 37%
need for more effective approaches for older adults.116

336 2020 ASCO EDUCATIONAL BOOK | asco.org/edbook

Downloaded from ascopubs.org by 2.57.77.86 on May 19, 2020 from 002.057.077.086


Copyright © 2020 American Society of Clinical Oncology. All rights reserved.
Recent Advances in Managing Acute Lymphoblastic Leukemia

The second-generation TKIs dasatinib and nilotinib (63% vs. 34%) compared with hyper-CVAD.125 A phase III
have been combined with chemotherapy, with EFS and trial of mini-hyper-CVD with or without inotuzumab is planned.
OS rates in the range of 42% to 55% and 46% to 72%,
Blinatumomab, an anti-CD19 bispecific T-cell engaging
respectively.114,116,120-122 Finally, a phase II trial evaluated
antibody, has been FDA approved for R-R B-ALL and has
the third-generation TKI ponatinib with hyper-CVAD.36,98
also demonstrated superiority to standard chemotherapy.59
This trial enrolled 66 patients, with 20 patients younger
This drug’s unique toxicity profile has included CRS and
than age 60. The rates of CR and complete molecular re-
neurotoxicity reminiscent of that seen with CAR T cells but at
mission were 100% and 77%, respectively, with a 3-year OS
a much lower incidence. In a phase II trial (SWOG1318),
of 77%. These results are impressive and demonstrated
patients at least age 65 with newly diagnosed Philadelphia-
increased EFS and OS compared with hyper-CVAD and
negative B-ALL were treated with blinatumomab for in-
other TKI-based therapies in a propensity analysis.98,123 This
duction, then received postremission therapy, followed by
analysis, in addition to the single-agent data, suggests
maintenance therapy with prednisone, vincristine, metho-
improved outcomes with successive-generation TKIs, al-
trexate, and mercaptopurine. Twenty-nine eligible patients
though no randomized trials have been reported. In addi-
were treated (median age, 75).126 The median bone marrow
tion, among patients with deep molecular remissions, there
blast count was 86.5%, and 34% of patients had poor-risk
were long-term survivors without ASCT. However, the dose
cytogenetics. The rate of response (CR and CR with in-
intensity of ponatinib correlated with adverse events, and
complete count recovery) was 66%, with no early deaths. At
the trial was amended for risk-adapted dosing, because
a median follow-up of 1.3 years, the estimated 1-year
three of 39 patients had a myocardial infarction at the
disease-free survival and OS were 58% and 67%, re-
continuous dose of 45 mg.36 Given the potential cardio-
spectively.126 Further follow-up will be needed to determine
vascular risks of ponatinib and the intensive nature of hyper-
the durability of these responses. A second clinical trial,
CVAD, this regimen is suitable only for truly fit older patients;
A041703, is sequencing inotuzumab and blinatumomab
strict organ function criteria were included in this trial.
treatment.
Given the short duration of remissions with TKIs alone and
For fit older adults, a recent U.S. Intergroup trial (ECOG
toxicities when combining with IC, other approaches are
1910) completed accrual (ages 30–70). Patients received
being evaluated, including combining dasatinib with im-
an intensive induction and postremission therapy, which
munotherapy, such as blinatumomab (SWOG 1318,
was modified based on age. There was a random assign-
NCT02143414), or combining various TKIs with low-
ment for patients who were MRD negative to either continue
dose chemotherapy.
standard therapy or receive blinatumomab followed by
Ongoing and Recent Clinical Trials for Patients With further chemotherapy. The results of this trial are eagerly
Philadelphia-Negative ALL awaited.

In consideration of the toxicities with standard IC, many Finally, the Bcl-2 inhibitor venetoclax is being evaluated in
current trials are reducing chemotherapy and adding novel both B-ALL and T-ALL. Bcl-2 is overexpressed in ALL.127 An
agents. Inotuzumab, an anti-CD22 antibody conjugated to ongoing trial evaluated the Bcl-2 inhibitor navitoclax plus
the cytotoxic agent calicheamicin, demonstrated higher venetoclax in combination with chemotherapy in R-R ALL.
rates of CR, MRD, and OS compared with standard che- Despite a heavily pretreated population, the response rate
motherapy in patients with R-R B-ALL due for salvage was encouraging at 56%.128 Another trial is evaluating
chemotherapy.60 This led to FDA approval of inotuzumab for venetoclax in combination with low-intensity chemotherapy
the treatment of R-R B-ALL. The drug is generally well for older patients with newly diagnosed ALL.129
tolerated, with a unique toxicity of hepatic VOD. Sub-
sequently, inotuzumab was combined with low-intensity Future Directions for Older Patients With ALL
chemotherapy (mini-hyper-CVD) for patients with Philadelphia- Multiple novel agents are being evaluated in Philadelphia-
negative B-ALL who were at least age 60.124 Fifty-two positive and -negative ALL. Incorporation of these agents
patients were treated (median age, 68). The incidence of with less-intensive chemotherapy is likely to improve out-
VOD was 8%, with a 12% incidence of treatment-related comes of older adults, and clinical trials are evaluating these
deaths.124 At a median follow-up of 29 months, the 2-year approaches. The presence of Philadelphia-like alterations
progression-free survival was encouraging at 59%.124 In and MLL rearrangements may make TKI-based therapy and
a propensity analysis comparing mini-hyper-CVD/inotuzumab MLL inhibitors in combination with low-dose therapy an-
with or without blinatumomab with hyper-CVAD, the anti- other option for these patients. Although CAR T cells are not
body plus low-intensity treatment demonstrated a higher yet FDA approved for adults older than age 26, these agents
response rate, lower rates of early death, lower rates of death are being used in clinical trials; as their toxicities are opti-
in remission, and higher 3-year EFS (64% vs. 34%) and OS mized, they are likely to become an option for older adults,

2020 ASCO EDUCATIONAL BOOK | asco.org/edbook 337

Downloaded from ascopubs.org by 2.57.77.86 on May 19, 2020 from 002.057.077.086


Copyright © 2020 American Society of Clinical Oncology. All rights reserved.
DeAngelo, Jabbour, and Advani

given their impressive clinical activity.83 The use of MRD to increasing availability of donors, transplantation is becoming
guide therapy will probably be incorporated into future trials, another option for patients who previously would not have
particularly because blinatumomab is now FDA approved in been candidates. Recent studies demonstrate encouraging
the MRD-positive setting. Finally, as outcomes with results for appropriate “elderly” candidates.130
reduced-intensity ASCT are improving and there is an

AFFILIATIONS CORRESPONDING AUTHOR


1
Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Daniel J. DeAngelo, MD, PhD, Dana-Farber Cancer Institute, 450
MA Brookline Ave., Boston, MA 02215; email: daniel_deangelo@dfci.
2
The University of Texas MD Anderson Cancer Center, Houston, TX harvard.edu.
3
Cleveland Clinic Taussig Cancer Institute, Cleveland, OH
AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST
AND DATA AVAILABILITY STATEMENT
Disclosures provided by the authors and data availability statement (if
applicable) are available with this article at DOI https://doi.org/10.1200/
EDBK_280175.

REFERENCES
1. National Cancer Institute Surveillance, Epidemiology, and End Results Program. Cancer Stat Facts: Leukemia - Acute Lymphocytic Leukemia (ALL). https://
seer.cancer.gov/statfacts/html/alyl.html. Accessed February 10, 2020.
2. Sant M, Allemani C, Tereanu C, et al; HAEMACARE Working Group. Incidence of hematologic malignancies in Europe by morphologic subtype: results of the
HAEMACARE project. Blood. 2010;116:3724-3734.
3. Coustan-Smith E, Gajjar A, Hijiya N, et al. Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia after first relapse.
Leukemia. 2004;18:499-504.
4. Gökbuget N, Kneba M, Raff T, et al; German Multicenter Study Group for Adult Acute Lymphoblastic Leukemia. Adult patients with acute lymphoblastic
leukemia and molecular failure display a poor prognosis and are candidates for stem cell transplantation and targeted therapies. Blood. 2012;120:1868-1876.
5. Pui CH, Relling MV, Downing JR. Acute lymphoblastic leukemia. N Engl J Med. 2004;350:1535-1548.
6. Pui CH, Campana D, Pei D, et al. Treating childhood acute lymphoblastic leukemia without cranial irradiation. N Engl J Med. 2009;360:2730-2741.
7. Larsen EC, Devidas M, Chen S, et al. Dexamethasone and high-dose methotrexate improve outcome for children and young adults with high-risk B-acute
lymphoblastic leukemia: a report from Children’s Oncology Group study AALL0232. J Clin Oncol. 2016;34:2380-2388.
8. Kantarjian H, Thomas D, O’Brien S, et al. Long-term follow-up results of hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone
(Hyper-CVAD), a dose-intensive regimen, in adult acute lymphocytic leukemia. Cancer. 2004;101:2788-2801.
9. Larson RA, Dodge RK, Linker CA, et al. A randomized controlled trial of filgrastim during remission induction and consolidation chemotherapy for adults with
acute lymphoblastic leukemia: CALGB study 9111. Blood. 1998;92:1556-1564.
10. DeAngelo DJ, Silverman LB, Couban S, et al. A multicenter phase II study using a dose intensified pediatric regimen in adults with untreated acute lymphoblastic
leukemia. Blood. 2006;108 (abstr 1858).
11. DeAngelo DJ, Stevenson KE, Dahlberg SE, et al. Long-term outcome of a pediatric-inspired regimen used for adults aged 18-50 years with newly diagnosed
acute lymphoblastic leukemia. Leukemia. 2015;29:526-534.
12. Huguet F, Leguay T, Raffoux E, et al. Pediatric-inspired therapy in adults with Philadelphia chromosome–negative acute lymphoblastic leukemia: the GRAALL-
2003 study [published correction appears in J Clin Oncol. 2009;27(15):2574]. J Clin Oncol. 2009;27:911-918.
13. Stock W, Luger SM, Advani AS, et al. A pediatric regimen for older adolescents and young adults with acute lymphoblastic leukemia: results of CALGB 10403.
Blood. 2019;133:1548-1559.
14. Fielding AK, Richards SM, Chopra R, et al; Eastern Cooperative Oncology Group. Outcome of 609 adults after relapse of acute lymphoblastic leukemia (ALL); an
MRC UKALL12/ECOG 2993 study. Blood. 2007;109:944-950.
15. Thomas DA, Kantarjian H, Smith TL, et al. Primary refractory and relapsed adult acute lymphoblastic leukemia: characteristics, treatment results, and prognosis
with salvage therapy. Cancer. 1999;86:1216-1230.
16. Berry DA, Zhou S, Higley H, et al. Association of minimal residual disease with clinical outcome in pediatric and adult acute lymphoblastic leukemia: a meta-
analysis. JAMA Oncol. 2017;3:e170580.
17. van Dongen JJ, van der Velden VH, Brüggemann M, et al. Minimal residual disease diagnostics in acute lymphoblastic leukemia: need for sensitive, fast, and
standardized technologies. Blood. 2015;125:3996-4009.
18. Gökbuget N, Hoelzer D. Treatment of adult acute lymphoblastic leukemia. Semin Hematol. 2009;46:64-75.
19. Hoelzer D, Thiel E, Löffler H, et al. Prognostic factors in a multicenter study for treatment of acute lymphoblastic leukemia in adults. Blood. 1988;71:123-131.

338 2020 ASCO EDUCATIONAL BOOK | asco.org/edbook

Downloaded from ascopubs.org by 2.57.77.86 on May 19, 2020 from 002.057.077.086


Copyright © 2020 American Society of Clinical Oncology. All rights reserved.
Recent Advances in Managing Acute Lymphoblastic Leukemia

20. Rowe JM, Buck G, Burnett AK, et al; MRC/NCRI Adult Leukemia Working Party. Induction therapy for adults with acute lymphoblastic leukemia: results of more
than 1500 patients from the international ALL trial: MRC UKALL XII/ECOG E2993. Blood. 2005;106:3760-3767.
21. Rowe JM. Prognostic factors in adult acute lymphoblastic leukaemia. Br J Haematol. 2010;150:389-405.
22. Maury S, Huguet F, Leguay T, et al; Group for Research on Adult Acute Lymphoblastic Leukemia. Adverse prognostic significance of CD20 expression in adults
with Philadelphia chromosome–negative B-cell precursor acute lymphoblastic leukemia. Haematologica. 2010;95:324-328.
23. Thomas DA, O’Brien S, Jorgensen JL, et al. Prognostic significance of CD20 expression in adults with de novo precursor B-lineage acute lymphoblastic
leukemia. Blood. 2009;113:6330-6337.
24. Maury S, Chevret S, Thomas X, et al; for GRAALL. Rituximab in B-Lineage adult acute lymphoblastic leukemia. N Engl J Med. 2016;375:1044-1053.
25. Marks DI, Paietta EM, Moorman AV, et al. T-cell acute lymphoblastic leukemia in adults: clinical features, immunophenotype, cytogenetics, and outcome from
the large randomized prospective trial (UKALL XII/ECOG 2993). Blood. 2009;114:5136-5145.
26. Coustan-Smith E, Mullighan CG, Onciu M, et al. Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia. Lancet Oncol.
2009;10:147-156.
27. Zhang J, Ding L, Holmfeldt L, et al. The genetic basis of early T-cell precursor acute lymphoblastic leukaemia. Nature. 2012;481:157-163.
28. Jain N, Lamb AV, O’Brien S, et al. Early T-cell precursor acute lymphoblastic leukemia/lymphoma (ETP-ALL/LBL) in adolescents and adults: a high-risk subtype.
Blood. 2016;127:1863-1869.
29. Dunsmore KP, Winter S, Devidas M, et al. COG AALL0434: a randomized trial testing nelarabine in newly diagnosed T-cell malignancy. J Clin Oncol. 2018;36
(suppl; abstr 10500).
30. Moorman AV, Ensor HM, Richards SM, et al. Prognostic effect of chromosomal abnormalities in childhood B-cell precursor acute lymphoblastic leukaemia:
results from the UK Medical Research Council ALL97/99 randomised trial. Lancet Oncol. 2010;11:429-438.
31. Aguiar RC, Sohal J, van Rhee F, et al; M.R.C. Adult Leukaemia Working Party. TEL-AML1 fusion in acute lymphoblastic leukaemia of adults. Br J Haematol.
1996;95:673-677.
32. Iacobucci I, Mullighan CG. Genetic basis of acute lymphoblastic leukemia. J Clin Oncol. 2017;35:975-983.
33. Foà R, Vitale A, Vignetti M, et al; GIMEMA Acute Leukemia Working Party. Dasatinib as first-line treatment for adult patients with Philadelphia
chromosome–positive acute lymphoblastic leukemia. Blood. 2011;118:6521-6528.
34. Vignetti M, Fazi P, Cimino G, 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’Adulto (GIMEMA)
LAL0201-B protocol. Blood. 2007;109:3676-3678.
35. Chalandon Y, Thomas X, Hayette S, et al; Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL). Randomized study of reduced-intensity
chemotherapy combined with imatinib in adults with Ph-positive acute lymphoblastic leukemia [published correction appears in Blood. 2015;126:1261].
Blood. 2015;125:3711-3719.
36. Short NJ, Kantarjian HM, Ravandi F, et al. Frontline hyper-CVAD plus ponatinib for patients with Philadelphia chromosome–positive acute lymphoblastic
leukemia: updated results of a phase II study. J Clin Oncol. 2017;35 (suppl; abstr 7013).
37. Moorman AV, Harrison CJ, Buck GA, et al; Adult Leukaemia Working Party, Medical Research Council/National Cancer Research Institute. Karyotype is an
independent prognostic factor in adult acute lymphoblastic leukemia (ALL): analysis of cytogenetic data from patients treated on the Medical Research Council
(MRC) UKALLXII/Eastern Cooperative Oncology Group (ECOG) 2993 trial. Blood. 2007;109:3189-3197.
38. Armstrong SA, Staunton JE, Silverman LB, et al. MLL translocations specify a distinct gene expression profile that distinguishes a unique leukemia. Nat Genet.
2002;30:41-47.
39. Stein EM, Garcia-Manero G, Rizzieri DA, et al. The DOT1L inhibitor pinometostat reduces H3K79 methylation and has modest clinical activity in adult acute
leukemia. Blood. 2018;131:2661-2669.
40. Armstrong SA, Mabon ME, Silverman LB, et al. FLT3 mutations in childhood acute lymphoblastic leukemia. Blood. 2004;103:3544-3546.
41. Holmfeldt L, Wei L, Diaz-Flores E, et al. The genomic landscape of hypodiploid acute lymphoblastic leukemia. Nat Genet. 2013;45:242-252.
42. Herold T, Schneider S, Metzeler KH, et al. Adults with Philadelphia chromosome–like acute lymphoblastic leukemia frequently have IGH-CRLF2 and JAK2
mutations, persistence of minimal residual disease and poor prognosis. Haematologica. 2017;102:130-138.
43. Roberts KG, Gu Z, Payne-Turner D, et al. High frequency and poor outcome of Philadelphia chromosome–like acute lymphoblastic leukemia in adults. J Clin
Oncol. 2017;35:394-401.
44. Roberts KG, Li Y, Payne-Turner D, et al. Targetable kinase-activating lesions in Ph-like acute lymphoblastic leukemia. N Engl J Med. 2014;371:1005-1015.
45. Roberts KG, Pei D, Campana D, et al. Outcomes of children with BCR-ABL1–like acute lymphoblastic leukemia treated with risk-directed therapy based on the
levels of minimal residual disease. J Clin Oncol. 2014;32:3012-3020.
46. Mullighan C, Downing J. Ikaros and acute leukemia. Leuk Lymphoma. 2008;49:847-849.
47. Mullighan CG, Miller CB, Radtke I, et al. BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros. Nature. 2008;453:110-114.
48. Zweidler-McKay PA, DeAngelo DJ, Douer D, et al. The safety and activity of BMS-906024, a gamma secretase inhibitor (GSI) with anti-notch activity, in patients
with relapsed T-cell acute lymphoblastic leukemia (T-ALL): initial results of a phase 1 trial. Blood. 2014;124:968.

2020 ASCO EDUCATIONAL BOOK | asco.org/edbook 339

Downloaded from ascopubs.org by 2.57.77.86 on May 19, 2020 from 002.057.077.086


Copyright © 2020 American Society of Clinical Oncology. All rights reserved.
DeAngelo, Jabbour, and Advani

49. Bassan R. Using minimal residual disease to improve treatment response definitions and hematopoietic cell transplantation strategy in acute leukemia. J Clin
Oncol. 2016;34:300-302.
50. Brüggemann M, Raff T, Kneba M. Has MRD monitoring superseded other prognostic factors in adult ALL? Blood. 2012;120:4470-4481.
51. Bassan R, Spinelli O, Oldani E, et al. Different molecular levels of post-induction minimal residual disease may predict hematopoietic stem cell transplantation
outcome in adult Philadelphia-negative acute lymphoblastic leukemia. Blood Cancer J. 2014;4:e225.
52. Dhédin N, Huynh A, Maury S, et al; GRAALL group. Role of allogeneic stem cell transplantation in adult patients with Ph-negative acute lymphoblastic leukemia.
Blood. 2015;125:2486-2496, quiz 2586.
53. Brüggemann M, Raff T, Flohr T, et al; German Multicenter Study Group for Adult Acute Lymphoblastic Leukemia. Clinical significance of minimal residual
disease quantification in adult patients with standard-risk acute lymphoblastic leukemia. Blood. 2006;107:1116-1123.
54. Spinelli O, Peruta B, Tosi M, et al. Clearance of minimal residual disease after allogeneic stem cell transplantation and the prediction of the clinical outcome of
adult patients with high-risk acute lymphoblastic leukemia. Haematologica. 2007;92:612-618.
55. Bassan R, Spinelli O, Oldani E, et al. Improved risk classification for risk-specific therapy based on the molecular study of minimal residual disease (MRD) in
adult acute lymphoblastic leukemia (ALL). Blood. 2009;113:4153-4162.
56. Ribera J-M, Oriol A, Morgades M, et al. Post-remission treatment with chemotherapy or allogeneic hematopoietic stem cell transplantation (alloHSCT) in adult
patients with high-risk (HR) Philadelphia chromosome–negative (Ph-neg) acute lymphoblastic leukemia (ALL) according to their minimal residual disease
(MRD). Final results of the Pethema ALL-HR-11 Trial. Blood. 2019;134 (suppl 1; abstr 826).
57. Jabbour E, Pui CH, Kantarjian H. Progress and innovations in the management of adult acute lymphoblastic leukemia. JAMA Oncol. 2018;4:1413-1420.
58. Gökbuget N, Dombret H, Ribera JM, et al. International reference analysis of outcomes in adults with B-precursor Ph-negative relapsed/refractory acute
lymphoblastic leukemia. Haematologica. 2016;101:1524-1533.
59. Kantarjian H, Stein A, Gökbuget N, et al. Blinatumomab versus chemotherapy for advanced acute lymphoblastic Leukemia. N Engl J Med. 2017;376:836-847.
60. Kantarjian HM, DeAngelo DJ, Stelljes M, et al. Inotuzumab ozogamicin versus standard therapy for acute lymphoblastic leukemia. N Engl J Med. 2016;375:740-753.
61. Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378:439-448.
62. Baeuerle PA, Reinhardt C. Bispecific T-cell engaging antibodies for cancer therapy. Cancer Res. 2009;69:4941-4944.
63. Bargou R, Leo E, Zugmaier G, et al. Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science. 2008;321:974-977.
64. Topp MS, Gökbuget N, Stein AS, 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.
65. Jabbour EJ, Gökbuget N, Kantarjian HM, et al. Transplantation in adults with relapsed/refractory acute lymphoblastic leukemia who are treated with bli-
natumomab from a phase 3 study. Cancer. 2019;125:4181-4192.
66. Brown PA, Ji L, Xu X, et al. A randomized phase 3 trial of blinatumomab vs. chemotherapy As post-reinduction therapy in high and intermediate risk (HR/IR) first
relapse of B-acute lymphoblastic Leukemia (B-ALL) in children and adolescents/Young adults (AYAs) demonstrates superior efficacy and tolerability of
blinatumomab: a report from Children’s Oncology Group study AALL1331. Blood. 2019;134 (suppl; abstr LBA-1).
67. Martinelli G, Boissel N, Chevallier P, 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.
68. Assi R, Kantarjian H, Short NJ, et al. Safety and efficacy of blinatumomab in combination with a tyrosine kinase inhibitor for the treatment of relapsed
Philadelphia chromosome–positive leukemia. Clin Lymphoma Myeloma Leuk. 2017;17:897-901.
69. Gökbuget N, Dombret H, Bonifacio M, et al. Blinatumomab for minimal residual disease in adults with B-cell precursor acute lymphoblastic leukemia [published
correction appears in Blood. 2019;133:2625]. Blood. 2018;131:1522-1531.
70. Gökbuget N, Dombret H, Giebel S, et al. Blinatumomab vs historic standard-of-care treatment for minimal residual disease in adults with B-cell precursor acute
lymphoblastic leukaemia. Eur J Haematol. 2020;104:299-309.
71. Raponi S, De Propris MS, Intoppa S, et al. Flow cytometric study of potential target antigens (CD19, CD20, CD22, CD33) for antibody-based immunotherapy in
acute lymphoblastic leukemia: analysis of 552 cases. Leuk Lymphoma. 2011;52:1098-1107.
72. Kantarjian H, Thomas D, Jorgensen J, et al. Inotuzumab ozogamicin, an anti-CD22-calecheamicin conjugate, for refractory and relapsed acute lymphocytic
leukaemia: a phase 2 study. Lancet Oncol. 2012;13:403-411.
73. Kantarjian H, Thomas D, Jorgensen J, et al. Results of inotuzumab ozogamicin, a CD22 monoclonal antibody, in refractory and relapsed acute lymphocytic
leukemia. Cancer. 2013;119:2728-2736.
74. DeAngelo DJ, Stock W, Stein AS, et al. Inotuzumab ozogamicin in adults with relapsed or refractory CD22-positive acute lymphoblastic leukemia: a phase 1/2
study. Blood Adv. 2017;1:1167-1180.
75. Kantarjian HM, DeAngelo DJ, Stelljes M, 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.
76. O’Brien MM. A phase 2 trial of inotuzumab ozogamicin (InO) in children and young adults with relapsed or refractory (R/R) CD22+ B-acute lymphoblastic
leukemia (B-ALL): results from Children’s Oncology Group Protocol AALL1621. Blood. 2019;134 (suppl 1; abstr 741).
77. Jabbour E, Ravandi F, Kebriaei P, et al. Salvage chemoimmunotherapy with inotuzumab ozogamicin combined with mini-hyper-CVD for patients with relapsed
or refractory Philadelphia chromosome–negative acute lymphoblastic leukemia: a phase 2 clinical trial. JAMA Oncol. 2018;4:230-234.

340 2020 ASCO EDUCATIONAL BOOK | asco.org/edbook

Downloaded from ascopubs.org by 2.57.77.86 on May 19, 2020 from 002.057.077.086


Copyright © 2020 American Society of Clinical Oncology. All rights reserved.
Recent Advances in Managing Acute Lymphoblastic Leukemia

78. Jabbour E, Gökbuget N, Advani A, 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.
79. Jabbour E, Sasaki K, Ravandi F, 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.
80. Jabbour E, Short NJ, Jorgensen JL, et al. Differential impact of minimal residual disease negativity according to the salvage status in patients with relapsed/
refractory B-cell acute lymphoblastic leukemia. Cancer. 2017;123:294-302.
81. Dombret H, Topp MS, Schuh AC, et al. Blinatumomab versus chemotherapy in first salvage or in later salvage for B-cell precursor acute lymphoblastic leukemia.
Leuk Lymphoma. 2019;60:2214-2222.
82. Grupp SA, Maude SL, Rives S, et al. Updated analysis of the efficacy and safety of tisagenlecleucel in pediatric and young adult patients with relapsed/refractory
(r/r) acute lymphoblastic leukemia. Blood. 2018;132:895.
83. Park JH, Rivière I, Gonen M, et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med. 2018;378:449-459.
84. Ghorashian S, Kramer AM, Onuoha S, 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.
85. Roddie C, O’Reilly MA, Pinto JDA, et al. AUTO1, a novel fast off CD19CAR delivers durable remissions and prolonged CAR T cell persistence with low CRS or
neurotoxicity in adult ALL. Blood. 2019;134 (suppl 1; abstr 226).
86. Fry TJ, Shah NN, Orentas RJ, et al. CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy. Nat
Med. 2018;24:20-28.
87. Schultz LM, Graham C, Yallop D, et al. Phase I trial using CD19/CD22 bispecific CAR T cells in pediatric and adult acute lymphoblastic leukemia (ALL). Blood.
2019;134 (suppl 1, abstr 744).
88. Benjamin R, Graham C, Yallop D, et al. Preliminary data on safety, cellular kinetics and anti-Leukemic activity of UCART19, an allogeneic anti-CD19 CAR T-cell
product, in a pool of adult and pediatric patients with high-risk CD19+ relapsed/refractory B-cell acute lymphoblastic leukemia. Blood. 2018;132 (suppl 1; abstr
896).
89. Benito JM, Godfrey L, Kojima K, et al. MLL-rearranged acute lymphoblastic leukemias activate BCL-2 through H3K79 methylation and are sensitive to the BCL-
2-specific antagonist ABT-199. Cell Rep. 2015;13:2715-2727.
90. Khaw SL, Suryani S, Evans K, et al. Venetoclax responses of pediatric ALL xenografts reveal sensitivity of MLL-rearranged leukemia. Blood. 2016;
128:1382-1395.
91. Frismantas V, Dobay MP, Rinaldi A, et al. Ex vivo drug response profiling detects recurrent sensitivity patterns in drug-resistant acute lymphoblastic leukemia.
Blood. 2017;129:e26-e37.
92. Lacayo NJ, Pullarkat VA, Stock W, et al. Safety and efficacy of venetoclax in combination with navitoclax in adult and pediatric relapsed/refractory acute
lymphoblastic leukemia and lymphoblastic lymphoma. Blood. 2019;134 (suppl 1; abstr 285).
93. Jain N, Stevenson KE, Winer ES, et al. A multicenter phase I study combining venetoclax with mini-hyper-CVD in older adults with untreated and relapsed/
refractory acute lymphoblastic leukemia. Blood. 2019. 134 (suppl 1; abstr 3867).
94. Chiaretti S, Foà R. Chemotherapy-free and reduced intensity approaches in elderly patients with B-lineage acute lymphoblastic leukemia. Eur J Intern Med.
2018;58:22-27.
95. Gökbuget N. Treatment of older patients with acute lymphoblastic leukemia. Hematology (Am Soc Hematol Educ Program). 2016;2016:573-579.
96. Chiaretti S, Vitale A, Cazzaniga G, et al. Clinico-biologic features of 5202 patients with acute lymphoblastic leukemia enrolled in the Italian AIEOP and GIMEMA
protocols and stratified in age cohorts. Haematologica. 2013;98:1702-1710.
97. Ribera JM, Garcı́a O, Fernández-Abellán P, et al; PETHEMA Group. Lack of negative impact of Philadelphia chromosome in older patients with acute
lymphoblastic leukaemia in the tyrosine kinase inhibitor era: comparison of two prospective parallel protocols. Br J Haematol. 2012;159:485-488.
98. Short NJ, Kantarjian H, Jabbour E, et al. Novel therapies for older adults with acute lymphoblastic leukemia. Curr Hematol Malig Rep. 2018;13:91-99.
99. Ribera J, Morgades M, Zamora L, et al; Spanish PETHEMA Group and the Spanish Society of Hematology. Prognostic significance of copy number alterations in
adolescent and adult patients with precursor B acute lymphoblastic leukemia enrolled in PETHEMA protocols. Cancer. 2015;121:3809-3817.
100. Khoral P, Atenafu EG, Craddock KJ, et al. Prognostic effect of complex karyotype, monosomal karyotype, and chromosome 17 abnormalities in B-cell acute
lymphoblastic leukemia. Clin Lymphoma Myeloma Leuk. 2017;17:215-219.
101. Issa GC, Kantarjian HM, Yin CC, et al. Prognostic impact of pretreatment cytogenetics in adult Philadelphia chromosome–negative acute lymphoblastic
leukemia in the era of minimal residual disease. Cancer. 2017;123:459-467.
102. Moorman AV, Chilton L, Wilkinson J, et al. A population-based cytogenetic study of adults with acute lymphoblastic leukemia. Blood. 2010;115:206-214.

103. Tasian SK, Hurtz C, Wertheim GB, et al. High incidence of Philadelphia chromosome–like acute lymphoblastic leukemia in older adults with B-ALL. Leukemia.
2017;31:981-984.
104. Thomas X, Olteanu N, Charrin C, et al. Acute lymphoblastic leukemia in the elderly: the Edouard Herriot Hospital experience. Am J Hematol. 2001;67:73-83.
105. Geyer MB, Hsu M, Devlin SM, et al. Overall survival among older US adults with ALL remains low despite modest improvement since 1980: SEER analysis.
Blood. 2017;129:1878-1881.

2020 ASCO EDUCATIONAL BOOK | asco.org/edbook 341

Downloaded from ascopubs.org by 2.57.77.86 on May 19, 2020 from 002.057.077.086


Copyright © 2020 American Society of Clinical Oncology. All rights reserved.
DeAngelo, Jabbour, and Advani

106. Goekbuget N, Beck J, Bruggeman M, et al. Moderate intensive chemotherapy including CNS prophylaxis with liposomal cytarabine is feasible and effective in
older patients with Ph-negative acute lymphoblastic leukemia (ALL): results of a prospective trial from the German Multicenter study group for adult ALL
(GMALL). Blood. 2012;120:1493.
107. Sive JI, Buck G, Fielding A, et al. Outcomes in older adults with acute lymphoblastic leukaemia (ALL): results from the international MRC UKALL XII/ ECOG 2993
trial. Br J Haematol. 2012;157:463-471.
108. O’Brien S, Thomas DA, Ravandi F, et al. Results of the hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone regimen in elderly
patients with acute lymphocytic leukemia. Cancer. 2008;113:2097-2101.
109. Larson RA, Dodge RK, Burns CP, et al. A five-drug remission induction regimen with intensive consolidation for adults with acute lymphoblastic leukemia:
cancer and leukemia group B study 8811. Blood. 1995;85:2025-2037.
110. Sancho JM, Ribera JM, Xicoy B, et al; PETHEMA Group. Results of the PETHEMA ALL-96 trial in elderly patients with Philadelphia chromosome–negative acute
lymphoblastic leukemia. Eur J Haematol. 2007;78:102-110.
111. Thomas DA, O’Brien S, Faderl S, et al. Chemoimmunotherapy with a modified hyper-CVAD and rituximab regimen improves outcome in de novo Philadelphia
chromosome–negative precursor B-lineage acute lymphoblastic leukemia. J Clin Oncol. 2010;28:3880-3889.
112. Ribera JM, Garcı́a O, Gil C, et al; PETHEMA Group. Comparison of intensive, pediatric-inspired therapy with non-intensive therapy in older adults aged 55-
65 years with Philadelphia chromosome–negative acute lymphoblastic leukemia. Leuk Res. 2018;68:79-84.
113. Ottmann OG, Wassmann B, Pfeifer H, et al; GMALL Study Group. Imatinib compared with chemotherapy as front-line treatment of elderly patients with
Philadelphia chromosome–positive acute lymphoblastic leukemia (Ph+ALL). Cancer. 2007;109:2068-2076.
114. Ravandi F, O’Brien SM, Cortes JE, et al. Long-term follow-up of a phase 2 study of chemotherapy plus dasatinib for the initial treatment of patients with
Philadelphia chromosome–positive acute lymphoblastic leukemia. Cancer. 2015;121:4158-4164.
115. Martinelli G, Piciocchi A, Papayannidis C, et al. First report of the Gimema LAL1811 phase 2 prospective study of the combination of steroids with ponatinib as
frontline therapy of elderly or unfit patients with Philadelphia chromosome–positive acute lymphoblastic leukemia. Blood. 2017; 130 (suppl 1; abstr 99).
116. Wieduwilt MJ. How should we treat older adults with Ph+ adult ALL and what novel approaches are being investigated? Best Pract Res Clin Haematol. 2017;
30:201-211.
117. Fielding AK, Rowe JM, Buck G, et al. UKALLXII/ECOG2993: addition of imatinib to a standard treatment regimen enhances long-term outcomes in Philadelphia-
positive acute lymphoblastic leukemia. Blood. 2014;123:843-850.
118. Bassan R, Rossi G, Pogliani EM, et al. Chemotherapy-phased imatinib pulses improve long-term outcome of adult patients with Philadelphia
chromosome–positive acute lymphoblastic leukemia: Northern Italy Leukemia Group protocol 09/00. J Clin Oncol. 2010;28:3644-3652.
119. Daver N, Thomas D, Ravandi F, et al. Final report of a phase II study of imatinib mesylate with hyper-CVAD for the front-line treatment of adult patients with
Philadelphia chromosome–positive acute lymphoblastic leukemia. Haematologica. 2015;100:653-661.
120. Ravandi F, O’Brien S, Thomas D, et al. First report of phase 2 study of dasatinib with hyper-CVAD for the frontline treatment of patients with Philadelphia
chromosome–positive (Ph+) acute lymphoblastic leukemia. Blood. 2010;116:2070-2077.
121. Kim DY, Joo YD, Lim SN, et al; Adult Acute Lymphoblastic Leukemia Working Party of the Korean Society of Hematology. Nilotinib combined with multiagent
chemotherapy for newly diagnosed Philadelphia-positive acute lymphoblastic leukemia. Blood. 2015;126:746-756.
122. Ravandi F, Othus M, O’Brien SM, et al. U.S. Intergroup study of chemotherapy plus dasatinib and allogeneic stem cell transplant in Philadelphia
chromosome–positive ALL. Blood Adv. 2016;1:250-259.
123. Sasaki K, Jabbour EJ, Ravandi F, et al. Hyper-CVAD plus ponatinib versus hyper-CVAD plus dasatinib as frontline therapy for patients with Philadelphia
chromosome–positive acute lymphoblastic leukemia: a propensity score analysis. Cancer. 2016;122:3650-3656.
124. Kantarjian H, Ravandi F, Short NJ, et al. Inotuzumab ozogamicin in combination with low-intensity chemotherapy for older patients with Philadelphia
chromosome–negative acute lymphoblastic leukaemia: a single-arm, phase 2 study. Lancet Oncol. 2018;19:240-248.
125. Jabbour EJ, Sasaki K, Ravandi F, et al. Inotuzumab ozogamicin in combination with low-intensity chemotherapy (mini-HCVD) with or without blinatumomab
versus standard intensive chemotherapy (HCVAD) as frontline therapy for older patients with Philadelphia chromosome–negative acute lymphoblastic leu-
kemia: a propensity score analysis. Cancer. 2019;125:2579-2586.
126. Advani AS, Moseley A, O’Dwyer KM, et al. Results of SWOG 1318: a phase 2 trial of blinatumomab followed by POMP (prednisone, vincristine, methotrexate, 6-
mercaptopurine) maintenance in elderly patients with newly diagnosed Philadelphia chromosome–negative B-cell acute lymphoblastic leukemia. Blood. 2018;
132 (suppl 1; abstr 33).
127. Alford SE, Kothari A, Loeff FC, et al. BH3 inhibitor sensitivity and bcl-2 dependence in primary acute lymphoblastic leukemia cells. Cancer Res. 2015;
75:1366-1375.
128. Lacayo N, Pullarkat V, Stock W, et al. Safety and efficacy of venetoclax in combination with navitoclax in adult and pediatric relapsed/refractory acute
lymphoblastic leukemia and lymphoblastic lymphoma. Blood. 2019;134 (suppl; abstr 285).
129. Jain N, Stevenson K, Winder ES, et al. A multicenter phase 1 study combining venetoclax with mini-hyper-CVD in older adults with untreated and relapsed/
refractory acute lymphoblastic leukemia. Blood. 2019 (abstr 3867).
130. Rosko A, Wang HL, de Lima M, et al. Reduced intensity conditioned allograft yields favorable survival for older adults with B-cell acute lymphoblastic leukemia.
Am J Hematol. 2017;92:42-49.

342 2020 ASCO EDUCATIONAL BOOK | asco.org/edbook

Downloaded from ascopubs.org by 2.57.77.86 on May 19, 2020 from 002.057.077.086


Copyright © 2020 American Society of Clinical Oncology. All rights reserved.

You might also like