By overcoming chemotherapeutic resistance, chimeric antigen receptor (CAR) T cells facilitate deep, complete remissions and offer the potential for long-term cure in a substantial fraction of patients with chemotherapy refractory disease. However, that success is tempered with 10% to 30% of patients not achieving remission and over half of patients treated eventually experiencing relapse. With over a decade of experience using CAR T cells in children, adolescents, and young adults (AYA) to treat relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL) and 5 years since the first US Food and Drug Administration approval, data defining the nuances of patient-specific risk factors are emerging. With the commercial availability of 2 unique CD19 CAR T-cell constructs for B-ALL, in this article, we review the current literature, outline our approach to patients, and discuss how individual factors inform strategies to optimize outcomes in children and AYA receiving CD19 CAR T cells. We include data from both prospective and recent large retrospective studies that offer insight into understanding when the risks of CAR T-cell therapy failure are high and offer perspectives suggesting when consolidative hematopoietic cell transplantation or experimental CAR T-cell and/or alternative immunotherapy should be considered. We also propose areas where prospective trials addressing the optimal use of CAR T-cell therapy are needed.

The first child to receive CD19-targeted chimeric antigen receptor (CAR) T cells for B-cell acute lymphoblastic leukemia (B-ALL) recently celebrated an important milestone: a decade in remission, vividly demonstrating that long-term, durable remissions can occur in highly chemotherapy refractory patients.1,2 There are currently 2 CD19 CAR T-cell constructs approved by the US Food and Drug Administration (FDA) for relapsed/refractory (r/r) B-ALL. Tisagenlecleucel (Kymriah), which incorporates the 4-1BB costimulatory domain, received FDA approval in 2017 for children and young adults ≤25 years,3,4 and brexucabtagene autoleucel (Tecartus), which incorporates the CD28 costimulatory domain, received FDA approval in 2021 for those ≥18 years with r/r B-ALL.5,6 

Emerging data encompassing both commercial and academic CAR T-cell constructs has led to the identification of risk factors for both response and long-term remission. These data can inform how we approach individual patients to optimize outcomes. Here, we describe in detail pre- and postinfusion risk factors associated with (1) nonresponse, (2) post-CAR relapse and event free-survival (EFS), and (3) long-term durable remission. We then propose a framework incorporating these risk factors to optimize CD19 CAR T-cell outcomes in children and adolescents and young adults, recognizing that rigorous trials are needed to validate our approaches. Through an evolving patient case study, we illustrate key points in the therapeutic path associated with CD19 CAR T-cell therapy.

Case: a 19-year-old male presents with first relapse of B-ALL 6 months after completion of therapy. At diagnosis, he was high-risk by age with normal cytogenetics and no molecularly targetable lesion, which did not change with relapse. He is treated with 4-drug reinduction, followed by blinatumomab, but has persistent CD19+ disease. Plans are made to proceed with CD19 CAR T cells. HLA typing is initiated to identify potential donors, but the family expresses a desire to avoid an allogeneic hematopoietic stem cell transplantation (HSCT).

Construct selection

Given this patient’s age and nonresponse to 2 lines of post-relapse therapy, he meets the indications to receive either of the 2 commercially available CAR T-cell constructs. Without head-to-head comparisons of tisagenlecleucel to brexucabtagene autoleucel and limited “real-world” experience with the latter, data comparing long-term outcomes for those from 18 to 25 years who are eligible for both are not available. The ZUMA-3 registration study for brexucabtagene autoleucel had a median age of 40 years (interquartile range, 28-52 years), limiting our ability to assess use in younger adults.6 The pediatric study (≤21 years) of brexucabtagene autoleucel is ongoing (NCT02625480, ZUMA-4).7 

With the above limitations, a key factor in construct selection is based on CAR T-cell persistence and treatment goals. Although tisagenlecleucel has established persistence and demonstrated the potential for durable remission without HSCT,8 the shorter persistence of CD28-containing CD19 CAR T-cell constructs (brexucabtagene autoleucel and its predecessor axicabtagene autoleucel9) and poor long-term survival without consolidative HSCT in this age group are critical considerations. We recommend consolidative HSCT for those in remission after receiving a CD28 CAR T-cell construct, particularly in HSCT-naïve patients.10 Thus, for families wishing to avoid or who are ineligible for HSCT, tisagenlecleucel would be preferable. Longitudinal data, preferably a comparative trial, would be needed to compare outcomes of tisagenlecleucel with brexucabtagene autoleucel if CAR T-cell therapy was used as a planned bridge to HSCT.

In addition, although refractory disease after relapse is an indication for either construct, the FDA label for tisagenlecleucel11 is restricted to second or later relapse, because the global registration ELIANA trial (NCT02435849) only included first relapse patients who were refractory to reinduction.4 Although the label for brexucabtagene autoleucel states that it is for adults with relapsed or refractory B-ALL,12 the international, multicenter phase 1 to 2 ZUMA-3 registration trial (NCT02614066) enrolled patients in first relapse only if the initial remission was <12 months.6,13 It should be noted that current therapies are insufficient for younger patients with a high- or intermediate-risk first relapse (eg, <36 months after diagnosis or late first relapse with minimal residual disease [MRD] positivity after reinduction). On the most recent phase 3 Children’s Oncology Group trial for first relapse of B-ALL (AALL1331), the 24-month intent-to-treat EFS was only 25% for those with a high-risk first relapse.14 The optimal use of CD19 CAR T cells in pediatric patients with a high- or intermediate-risk first relapse needs to be further explored, as it may lead to better outcomes than current approaches.

Previous blinatumomab use

Our patient received the CD3-CD19 bispecific T-cell engager blinatumomab, which is FDA-approved for the treatment of r/r B-ALL.15,16 Because of the concern that CD19 modulation with blinatumomab could impair subsequent CD19 CAR T-cell outcomes,17-19 the global registration trial of tisagenlecleucel excluded blinatumomab-exposed patients.4 Therefore, until recently, there was minimal data to shed light on the impact of sequential CD19-directed immunotherapy.

To address this gap, the association of prior blinatumomab exposure with CD19 CAR T-cell outcomes was assessed among 420 patients from 7 pediatric centers through the CAR-multicenter analysis (CAR-MA).20 The analysis revealed that blinatumomab exposure alone was not a risk factor for worse outcomes. However, nonresponse to prior blinatumomab was associated with lower complete remission (CR) rates (65% vs 93%) and shorter EFS, relapse-free survival (RFS), and overall survival (OS) (Table 1), with relapse rates exceeding 50% by 6 months (Figure 1A). These results suggest that blinatumomab nonresponse may serve as a proxy for identifying patients with either intrinsic resistance to CD19-targeting, T-cell dysfunction, or other high-risk features.20 However, use of blinatumomab and/or nonresponse should not be a contraindication to the use of CAR T cells, particularly as most patients refractory to blinatumomab do achieve a CR with CAR T-cell therapy.

Table 1.

Risk factors for inferior outcomes after CD19 CAR T cells in pediatric B-ALL

Risk factorSummarySpecific data
Baseline disease characteristics 
Leukemia cytogenetics Overall, there is not a clear impact of leukemia cytogenetic lesions on CD19 CAR outcomes. However, several rare, specific lesions may deserve additional consideration:
1. KMT2Ar: similar relapse risk to other groups, but higher incidence of myeloid lineage switch with dismal survival after lineage switch leading to worse OS.
2. TP53: associated with substantially worse LFS and OS in a small sample.
3. Hypodiploid: poor outcomes reported in several small studies. 
CHOP study21 (N = 231): similar RFS for patients with high-, intermediate-, and low-risk cytogenetics. Patients with KMT2Ar infant B-ALL (n = 13) had similar RFS to other groups, but worse OS (HR 3.6). Of 8 patients with hypodiploid B-ALL, 5 relapsed.
Robert Debre and Saint Louis University Hospitals study22 (N = 51): high-risk genetic lesions were not associated with CIR, EFS, or OS.
CAR-MA studies20,23 (N = 420): KMT2Ar (n = 38) associated with higher risk of lineage switch (HR 32.35) and lower median OS (25.3 vs 51.9 mo). EFS for patients with hypodiploid B-ALL (n = 14) was approximately 25% by 12 mo after CAR.
Lu Daopei Hospital phase 1/2 study24 (N = 110): patients with TP53 mutations had lower 6-mo LFS (42% vs 83%, P = .0002) and OS (52% vs 89%, P < .0001) than those without TP53 alterations. 
Molecular targets As above, CD19 CAR T cells are effective across multiple molecular subtypes of B-ALL. For patients with Philadelphia-positive or certain subtypes of Philadelphia-like B-ALL, the question has been raised of combining CAR T cells with the multi–tyrosine kinase inhibitor dasatinib. This is not currently recommended, as high-dose dasatinib is toxic to T cells. Preclinical dasatinib experiments:25 in a xenograft model, dasatinib rapidly and reversibly inhibited CAR T-cell activation and proliferation, cytokine production, and CAR T-cell killing. The authors suggested that dasatinib could potentially be repurposed as an “on” and “off” switch for CAR T cells. Further, the authors posited that intermittent or low-dose dasatinib may provide a synergistic therapeutic effect with CAR T cells. No human data exists, so this approach is not recommended. 
Previous therapy 
Blinatumomab Two single/dual-center analyses suggested that prior blinatumomab may affect response rates and relapse risk. A large, multicenter analysis showed that nonresponse to prior blinatumomab, rather than blinatumomab exposure alone, was associated with inferior response rates and EFS. CHOP study (N = 166): composite outcome of NR, CD19 MRD/relapse was more frequent in blinatumomab-exposed patients.26,
Robert Debre and Saint Louis University Hospitals study22 (N = 51): prior blinatumomab was associated with early CAR failure (P = .01), increased CIR (HR 2.6), and shorter EFS (HR 3.0) and OS (HR 5.5).
CAR-MA study (N = 420): blinatumomab nonresponders had lower CR rates to CD19 CAR T cells and worse 6-mo EFS (CR, 65%; EFS, 27%) than blinatumomab responders (CR, 93%; EFS, 67%) or blinatumomab-naïve patients (CR, 94%; EFS, 73%).20  
Quantity of previous therapy Two multicenter analyses showed that a greater quantity of prior therapy, possibly indicating more refractory disease, was associated with worse outcomes. PRWCC study (N = 185): a greater number of prior lines of therapy was associated with worse OS by multivariable analysis (HR 1.4, P = .022).27,
CAR-MA study23 (N = 420): at least 2 prior CRs were associated with a higher cumulative incidence of CD19-positive relapse by multivariable analysis (HR 1.3, P = .008). 
Factors at time of infusion 
Bone marrow disease burden Multiple clinical trials and multicenter analyses clearly demonstrate that high disease burden (HD burden), generally defined as ≥5% bone marrow blasts, is associated with increased risk of relapse, compared with low disease burden (LD burden). CHOP clinical trials: in a trial of tisagenlecleucel (N = 70),28 patients with HD burden (>40% blasts) had inferior 24-mo EFS (34% vs 78%) and OS (60% vs 92%) compared with LD burden. In a trial of humanized CD19 CAR (N = 74),29 HD burden was associated with inferior RFS.
CAR-MA studies20,23 (N = 420): HD burden (≥5% bone marrow blasts) was associated with inferior EFS, RFS, and OS. HD burden was independently associated with worse EFS (HR 2.5, P < .001) by multivariable analysis, and specifically associated with a higher cumulative incidence of CD19 relapse (HR 5.2, P < .001).
PRWCC study27 (N = 185): patients with HD burden (≥5% bone marrow blasts) had lower 12-mo EFS (31% vs 70%, P < .0001) and OS (58% vs 85%, P < .0001) compared with LD burden. HD burden was independently associated with OS by multivariable analysis (HR 5.1, P = .002).
St Jude and JHU study30 (N = 30): HD burden (≥5% bone marrow blasts) was independently associated with inferior EFS (HR 6.0, P = .038) and OS (HR 4.2, P = .015).
Robert Debre and Saint Louis University Hospitals study22 (N = 51): HD burden (≥1% bone marrow blasts) was associated with a higher cumulative incidence of CD19 relapse (SHR 10.4, P = .03) in a competing risks analysis. 
EMD Active non-CNS EMD at time of CD19 CAR infusion has been associated with worse EFS. Prior extramedullary or CNS disease, however, has not been associated with worse outcomes. CAR-MA study20 (N = 420): active non-CNS EMD at infusion was independently associated with worse EFS (HR 1.9, P = .01)
PRWCC study31 (N = 184): CNS and non-CNS EMD were not associated with RFS or OS differences. 
CD19+ antigen load Bone marrow CD19+ antigen load has been associated with CAR persistence, which, in turn, would be expected to affect relapse risk. Seattle PLAT-02 trial (N = 45): CD19+ antigen load measured as a combination of normal B cells and blasts <15% before lymphodepletion was independently associated with early loss of BCA (HR 3.0, P = .005).32  
Lymphodepletion Although most patients receive standard dosing of cyclophosphamide and fludarabine based on body surface area for lymphodepletion, this may not be the optimal regimen for all patients. In 2 recent retrospective studies, for example, suboptimal fludarabine exposure was associated with increased relapse risk and shorter persistence. In addition, bendamustine has been explored as an alternative lymphodepleting agent, especially in light of recent fludarabine shortages; however, there is no published data on its use in pediatric B-ALL. PRWCC study33 (N = 152): suboptimal fludarabine exposure, defined as AUC <13.8 mg × h/L and estimated by a validated population pharmacokinetic model, was associated with a higher CIR (HR 2.5, P = .005) and higher risk of a composite end point of relapse or loss of BCA (HR 2.0, P = .01) compared with optimal fludarabine exposure.
Princess Maxima study34 (N = 26): a cumulative fludarabine AUC <14 mg × h/L was associated with a higher frequency of CD19+ relapse within 1 y (100% vs 27%, P = .0001) and probability of losing BCA within 6 mo (77% vs 37%, P = .009) than AUC >14 mg × h/L.
Multicenter, adult large B-cell lymphoma study35 (N = 132): compared with fludarabine/cyclophosphamide lymphodepletion before tisagenlecleucel, bendamustine lymphodepletion demonstrated similar efficacy and lower rates of CRS, neurotoxicity, and hematologic toxicities. 
Postinfusion factors 
Short CAR persistence (loss of BCA) Short CAR persistence, as evidenced by loss of peripheral BCA, has been highly associated with increased risk of relapse. However, the duration of BCA needed to mitigate relapse risk has not been fully elucidated. Seattle PLAT-02 trial32 (N = 45): loss of BCA was associated with increase relapse risk (HR 3.5, P = .04).
Pooled ELIANA/ENSIGN analysis36 (N = 143): loss of BCA within 1 y was associated with increased relapse risk (HR 4.5, P < .001). Patients with loss of BCA within 6 mo had a 24-mo EFS of 14%.
CHOP humanized CD19 CAR T-cell trial29 (N = 74): when treated as a time-varying covariate, B-cell recovery was associated with worse RFS (P = .011). 
Detectable NGS- MRD or IG/TCR RQ-PCR MRD In a large, retrospective analysis, any detectable NGS-MRD at different time points after CD19 CAR T cells was highly predictive of subsequent relapse. In another retrospective analysis, detectable IG/TCR MRD (>10−4) at day 28 was highly predictive of subsequent relapse. Pooled ELIANA/ENSIGN analysis36 (N = 143): NGS-MRD >0 at day 28 (HR 4.9, P < .001) and month 3 (HR 12, P < .001) after CD19 CAR T cells were highly and independently associated with relapse.
Robert Debre and Saint Louis University Hospitals study22 (N=51): detectable IG/TCR RQ-PCR-MRD (>10−4) at day 28 after CD19 CAR T cells was highly and independently associated with relapse (HR 6.0, P = .006). 
Risk factorSummarySpecific data
Baseline disease characteristics 
Leukemia cytogenetics Overall, there is not a clear impact of leukemia cytogenetic lesions on CD19 CAR outcomes. However, several rare, specific lesions may deserve additional consideration:
1. KMT2Ar: similar relapse risk to other groups, but higher incidence of myeloid lineage switch with dismal survival after lineage switch leading to worse OS.
2. TP53: associated with substantially worse LFS and OS in a small sample.
3. Hypodiploid: poor outcomes reported in several small studies. 
CHOP study21 (N = 231): similar RFS for patients with high-, intermediate-, and low-risk cytogenetics. Patients with KMT2Ar infant B-ALL (n = 13) had similar RFS to other groups, but worse OS (HR 3.6). Of 8 patients with hypodiploid B-ALL, 5 relapsed.
Robert Debre and Saint Louis University Hospitals study22 (N = 51): high-risk genetic lesions were not associated with CIR, EFS, or OS.
CAR-MA studies20,23 (N = 420): KMT2Ar (n = 38) associated with higher risk of lineage switch (HR 32.35) and lower median OS (25.3 vs 51.9 mo). EFS for patients with hypodiploid B-ALL (n = 14) was approximately 25% by 12 mo after CAR.
Lu Daopei Hospital phase 1/2 study24 (N = 110): patients with TP53 mutations had lower 6-mo LFS (42% vs 83%, P = .0002) and OS (52% vs 89%, P < .0001) than those without TP53 alterations. 
Molecular targets As above, CD19 CAR T cells are effective across multiple molecular subtypes of B-ALL. For patients with Philadelphia-positive or certain subtypes of Philadelphia-like B-ALL, the question has been raised of combining CAR T cells with the multi–tyrosine kinase inhibitor dasatinib. This is not currently recommended, as high-dose dasatinib is toxic to T cells. Preclinical dasatinib experiments:25 in a xenograft model, dasatinib rapidly and reversibly inhibited CAR T-cell activation and proliferation, cytokine production, and CAR T-cell killing. The authors suggested that dasatinib could potentially be repurposed as an “on” and “off” switch for CAR T cells. Further, the authors posited that intermittent or low-dose dasatinib may provide a synergistic therapeutic effect with CAR T cells. No human data exists, so this approach is not recommended. 
Previous therapy 
Blinatumomab Two single/dual-center analyses suggested that prior blinatumomab may affect response rates and relapse risk. A large, multicenter analysis showed that nonresponse to prior blinatumomab, rather than blinatumomab exposure alone, was associated with inferior response rates and EFS. CHOP study (N = 166): composite outcome of NR, CD19 MRD/relapse was more frequent in blinatumomab-exposed patients.26,
Robert Debre and Saint Louis University Hospitals study22 (N = 51): prior blinatumomab was associated with early CAR failure (P = .01), increased CIR (HR 2.6), and shorter EFS (HR 3.0) and OS (HR 5.5).
CAR-MA study (N = 420): blinatumomab nonresponders had lower CR rates to CD19 CAR T cells and worse 6-mo EFS (CR, 65%; EFS, 27%) than blinatumomab responders (CR, 93%; EFS, 67%) or blinatumomab-naïve patients (CR, 94%; EFS, 73%).20  
Quantity of previous therapy Two multicenter analyses showed that a greater quantity of prior therapy, possibly indicating more refractory disease, was associated with worse outcomes. PRWCC study (N = 185): a greater number of prior lines of therapy was associated with worse OS by multivariable analysis (HR 1.4, P = .022).27,
CAR-MA study23 (N = 420): at least 2 prior CRs were associated with a higher cumulative incidence of CD19-positive relapse by multivariable analysis (HR 1.3, P = .008). 
Factors at time of infusion 
Bone marrow disease burden Multiple clinical trials and multicenter analyses clearly demonstrate that high disease burden (HD burden), generally defined as ≥5% bone marrow blasts, is associated with increased risk of relapse, compared with low disease burden (LD burden). CHOP clinical trials: in a trial of tisagenlecleucel (N = 70),28 patients with HD burden (>40% blasts) had inferior 24-mo EFS (34% vs 78%) and OS (60% vs 92%) compared with LD burden. In a trial of humanized CD19 CAR (N = 74),29 HD burden was associated with inferior RFS.
CAR-MA studies20,23 (N = 420): HD burden (≥5% bone marrow blasts) was associated with inferior EFS, RFS, and OS. HD burden was independently associated with worse EFS (HR 2.5, P < .001) by multivariable analysis, and specifically associated with a higher cumulative incidence of CD19 relapse (HR 5.2, P < .001).
PRWCC study27 (N = 185): patients with HD burden (≥5% bone marrow blasts) had lower 12-mo EFS (31% vs 70%, P < .0001) and OS (58% vs 85%, P < .0001) compared with LD burden. HD burden was independently associated with OS by multivariable analysis (HR 5.1, P = .002).
St Jude and JHU study30 (N = 30): HD burden (≥5% bone marrow blasts) was independently associated with inferior EFS (HR 6.0, P = .038) and OS (HR 4.2, P = .015).
Robert Debre and Saint Louis University Hospitals study22 (N = 51): HD burden (≥1% bone marrow blasts) was associated with a higher cumulative incidence of CD19 relapse (SHR 10.4, P = .03) in a competing risks analysis. 
EMD Active non-CNS EMD at time of CD19 CAR infusion has been associated with worse EFS. Prior extramedullary or CNS disease, however, has not been associated with worse outcomes. CAR-MA study20 (N = 420): active non-CNS EMD at infusion was independently associated with worse EFS (HR 1.9, P = .01)
PRWCC study31 (N = 184): CNS and non-CNS EMD were not associated with RFS or OS differences. 
CD19+ antigen load Bone marrow CD19+ antigen load has been associated with CAR persistence, which, in turn, would be expected to affect relapse risk. Seattle PLAT-02 trial (N = 45): CD19+ antigen load measured as a combination of normal B cells and blasts <15% before lymphodepletion was independently associated with early loss of BCA (HR 3.0, P = .005).32  
Lymphodepletion Although most patients receive standard dosing of cyclophosphamide and fludarabine based on body surface area for lymphodepletion, this may not be the optimal regimen for all patients. In 2 recent retrospective studies, for example, suboptimal fludarabine exposure was associated with increased relapse risk and shorter persistence. In addition, bendamustine has been explored as an alternative lymphodepleting agent, especially in light of recent fludarabine shortages; however, there is no published data on its use in pediatric B-ALL. PRWCC study33 (N = 152): suboptimal fludarabine exposure, defined as AUC <13.8 mg × h/L and estimated by a validated population pharmacokinetic model, was associated with a higher CIR (HR 2.5, P = .005) and higher risk of a composite end point of relapse or loss of BCA (HR 2.0, P = .01) compared with optimal fludarabine exposure.
Princess Maxima study34 (N = 26): a cumulative fludarabine AUC <14 mg × h/L was associated with a higher frequency of CD19+ relapse within 1 y (100% vs 27%, P = .0001) and probability of losing BCA within 6 mo (77% vs 37%, P = .009) than AUC >14 mg × h/L.
Multicenter, adult large B-cell lymphoma study35 (N = 132): compared with fludarabine/cyclophosphamide lymphodepletion before tisagenlecleucel, bendamustine lymphodepletion demonstrated similar efficacy and lower rates of CRS, neurotoxicity, and hematologic toxicities. 
Postinfusion factors 
Short CAR persistence (loss of BCA) Short CAR persistence, as evidenced by loss of peripheral BCA, has been highly associated with increased risk of relapse. However, the duration of BCA needed to mitigate relapse risk has not been fully elucidated. Seattle PLAT-02 trial32 (N = 45): loss of BCA was associated with increase relapse risk (HR 3.5, P = .04).
Pooled ELIANA/ENSIGN analysis36 (N = 143): loss of BCA within 1 y was associated with increased relapse risk (HR 4.5, P < .001). Patients with loss of BCA within 6 mo had a 24-mo EFS of 14%.
CHOP humanized CD19 CAR T-cell trial29 (N = 74): when treated as a time-varying covariate, B-cell recovery was associated with worse RFS (P = .011). 
Detectable NGS- MRD or IG/TCR RQ-PCR MRD In a large, retrospective analysis, any detectable NGS-MRD at different time points after CD19 CAR T cells was highly predictive of subsequent relapse. In another retrospective analysis, detectable IG/TCR MRD (>10−4) at day 28 was highly predictive of subsequent relapse. Pooled ELIANA/ENSIGN analysis36 (N = 143): NGS-MRD >0 at day 28 (HR 4.9, P < .001) and month 3 (HR 12, P < .001) after CD19 CAR T cells were highly and independently associated with relapse.
Robert Debre and Saint Louis University Hospitals study22 (N=51): detectable IG/TCR RQ-PCR-MRD (>10−4) at day 28 after CD19 CAR T cells was highly and independently associated with relapse (HR 6.0, P = .006). 

AUC, area under the curve; CHOP, Children’s Hospital of Philadelphia; CIR, cumulative incidence of relapse; JHU, Johns Hopkins University; NGS-MRD, detectable minimal residual disease by next-generation sequencing; IG/TCR RQ-PCR MRD, allele-specific real-time quantitative PCR of leukemia-specific immune-receptor gene rearrangements (either IG or TCR sequences); SHR, subdistribution hazard ratio; St Jude, St Jude Children’s Research Hospital; TCR, T-cell receptor.

Figure 1.

Impact of previous blinatumomab treatment and disease burden on EFS following CD19 CAR T cells and OS following relapse. EFS, defined as the time from CD19 CAR T-cell infusion to one of the following events: no response, relapse, or death. (A) EFS stratified by blinatumomab-naïve patients (no blina—teal) vs blinatumomab-exposed patients who achieved a CR to blinatumomab (blina-CR—blue) vs blinatumomab-exposed patients who did not achieve a CR to blinatumomab (blina-no CR—red). P values for EFS curves: .59 (no blina vs blina-CR); .01 (blina-CR vs blina-no CR); .001 (no blina vs blina-no CR). (B) EFS stratified by high disease burden (≥5% bone marrow blasts—blue [high]) vs low disease burden (<5% bone marrow blasts—red (low). (C) OS following relapse, stratified by relapse immunophenotype. Median OS for CD19+ relapse was 18.9 months (95% CI, 11.2-27.0 months). Median OS for CD19 relapse was 9.7 months (95% CI, 6.9-15.9 months). Median OS for LS was 3.7 months (95% CI, 1.2-7.0 months). Red: CD19+; blue: CD19; green: LS. CI, confidence interval; LS, lineage switch. (A-B) Reproduced with permission from Wolters Kluwer Health, license number 5363041439390, from Myers et al20; (C) reproduced with permission from Elsevier, license number 5363050768789, from Lamble et al.23 

Figure 1.

Impact of previous blinatumomab treatment and disease burden on EFS following CD19 CAR T cells and OS following relapse. EFS, defined as the time from CD19 CAR T-cell infusion to one of the following events: no response, relapse, or death. (A) EFS stratified by blinatumomab-naïve patients (no blina—teal) vs blinatumomab-exposed patients who achieved a CR to blinatumomab (blina-CR—blue) vs blinatumomab-exposed patients who did not achieve a CR to blinatumomab (blina-no CR—red). P values for EFS curves: .59 (no blina vs blina-CR); .01 (blina-CR vs blina-no CR); .001 (no blina vs blina-no CR). (B) EFS stratified by high disease burden (≥5% bone marrow blasts—blue [high]) vs low disease burden (<5% bone marrow blasts—red (low). (C) OS following relapse, stratified by relapse immunophenotype. Median OS for CD19+ relapse was 18.9 months (95% CI, 11.2-27.0 months). Median OS for CD19 relapse was 9.7 months (95% CI, 6.9-15.9 months). Median OS for LS was 3.7 months (95% CI, 1.2-7.0 months). Red: CD19+; blue: CD19; green: LS. CI, confidence interval; LS, lineage switch. (A-B) Reproduced with permission from Wolters Kluwer Health, license number 5363041439390, from Myers et al20; (C) reproduced with permission from Elsevier, license number 5363050768789, from Lamble et al.23 

Close modal

CD19 expression considerations

Although CD19 escape was not found to be the primary mechanism of inferior outcomes in blinatumomab-exposed patients who retained robust CD19 expression,20 for those whose CD19 expression became dim following blinatumomab, the risk of antigen-negative relapse after CD19 CAR T cells in the CAR-MA study was particularly high.20 Of note, the incidence of pediatric patients becoming fully CD19 following blinatumomab is unknown. Given the increasing use of blinatumomab, additional studies assessing CD19 expression over time and the effect of sequential CD19 targeting with different agents are warranted.

In addition, for blinatumomab-naïve patients, the risk of relapse with CD19 disease remains of concern, indeed in the ELIANA registration study, most relapses were CD19.4 Recent studies show an association between high disease burden and emergence of CD19 relapse.22,23 Whether this reflects an increased risk of a CD19 clone being present in a high disease burden scenario or whether high disease burden results in the persistence of functional CAR T cells and, over time, more genetic pressure for CD19 escape warrants future study.

Additional risk factors

Baseline patient characteristics, including age, sex, and history of trisomy 21, do not appear to affect outcomes.20,24,27,37 Surprisingly, leukemia cytogenetics also generally do not affect outcomes (Table 1). In a large, retrospective analysis from Children’s Hospital of Philadelphia (n = 231) patients with high-, intermediate-, and low-risk cytogenetics all had similar RFS, illustrating the remarkable ability of CAR T cells to overcome biological risk factors that portend a poor response to chemotherapy.38 A smaller study from 2 centers in France (n = 51) demonstrated similar results.22 

Several specific cytogenetic lesions, however, deserve additional consideration. Patients with KMT2A rearrangement (KMT2Ar) have similar RFS compared with other cytogenetic groups after CAR T-cell therapy; however, an increased risk of myeloid lineage switch has been noted (reported populations have been mostly infant ALL).23,38,39 Unfortunately, survival after a lineage switch is dismal, leading to an inferior OS. In addition, poor long-term outcomes have been reported in samples of patients with TP53 mutations24 or hypodiploid B-ALL,20,38 but individual cohorts have been too small to draw firm conclusions.

Finally, an increased quantity of prior therapy, reflective of more refractory disease and potentially dysfunctional T cells, has been associated with worse CD19 CAR outcomes. A Pediatric Real World CAR Consortium (PRWCC) study found that a greater number of lines of prior therapy (measured as a continuous variable) was independently associated with worse OS with a hazard ratio (HR) of 1.4 (P = .022).27 

Case (continued): given the desire to avoid allogeneic HSCT, the patient moved forward with tisagenlecleucel. With confirmed CD19+ expression following blinatumomab and an adequate lymphocyte count, plans were made to proceed with apheresis. However, with 26% of bone marrow blasts present, bridging therapy was deemed necessary.

Access

Despite the commercial availability of CAR T cells, there are many barriers to access, and making an early referral, obtaining insurance approval, and potentially transitioning a patient to another center can be time-intensive, leading to delays in therapy. Insurance approval must be obtained both for apheresis and CAR T-cell therapy. This can be especially challenging in situations where patients are at very high risk and centers attempt to preemptively collect before relapse, or when patients need to move across state or international borders for therapy. Early referral for CAR T-cell therapy may help address these issues and may also be beneficial for subgroups with chemotherapy refractory disease, allowing earlier collection of healthy T cells that could lead to better outcomes.40 

Leukapheresis

The first step of CD19 CAR T-cell manufacturing is the collection of autologous peripheral blood mononuclear cells by leukapheresis. Optimal collection practices may improve the outcome as T-cell quality affects CAR T-cell functionality. Compelling data demonstrate that T-cell populations enriched for early lineage cells have improved expansion and that early lineage cells are selectively depleted with continued courses of chemotherapy.40,41 Hence, if CD19 CAR T cells are being considered, we recommend collection as early as possible, before initiation of chemotherapy for relapse when feasible. In addition, preemptive apheresis may be considered for very high-risk patient subsets (eg, infant ALL MRD+ after induction).

The timing of apheresis involves consideration of multiple factors, including disease progression and time from previous therapy (Table 2). Manufacturing standards for tisagenlecleucel require at least 1 × 109 CD3+ cells, with rare exceptions for infants and very small children. Therefore, a minimum absolute lymphocyte count of ≥300 cells per μL or a CD3+ cell count of 100 to 200 cells per μL is recommended, with higher values preferable. Although there may be a narrow window for collection, collection is generally not advisable in patients with rapid disease progression and elevated circulating blasts (white blood cell > 20-40 000/μL), as high blast counts can interfere with manufacturing,42 although the threshold below which apheresis is acceptable has yet to be defined and may change with improvements in manufacturing and selection methodologies.43 Importantly, such patients can often be successfully collected after resolution of hyperleukocytosis with additional chemotherapy.

Table 2.

Considerations for apheresis timing and bridging therapy before CD19 CAR T-cell therapy

Treatment modalityTherapiesConsiderations
Timing to apheresis  The following factors should be taken into consideration to determine optimal timing for apheresis:
Quantity of peripheral T cells: a minimum absolute lymphocyte count of >300 cells per μL or CD3+ cell count of 100 to 200 cells per μL is recommended.
Circulating peripheral blasts: it is generally not advisable to collect with a high circulating blast count (WBC > 20-40 000/μL with >50%-80% leukemic blasts) because this can interfere with manufacturing.42
Quantity of prior therapy exposure: we recommend collecting T cells as early as feasible because continued courses of chemotherapy selectively deplete early lineage cells, which are important for T-cell expansion.40,41 
Time from prior therapy: a minimum treatment-free period is needed before collection, ranging from 72 h for corticosteroids, to 2 wk for cytotoxic chemotherapy, to longer periods for certain immunotherapies (eg, 6 wk-3 mo for pembrolizumab).
Patients with a history of HSCT: we recommend that apheresis occur at least 100 to 120 d after HSCT and immunosuppression be discontinued at least 4 wk before apheresis without signs of graft-versus-host disease to avoid the risk of collecting alloreactive T-cells. 
Chemotherapy Intensive regimens: 4-drug induction (vincristine, corticosteroid, asparaginase and anthracycline); cyclophosphamide with etoposide; high-dose cytarabine.
Less-intensive regimens: maintenance-style chemotherapy (oral mercaptopurine, oral methotrexate, ±vincristine, and short course of corticosteroids); abbreviated consolidation cycle with cyclophosphamide and low-dose cytarabine; intermediate-dose VP-16/cytosine arabinoside; intermediate-dose methotrexate; 3-drug induction (vincristine, corticosteroid, and asparaginase)
IT chemotherapy should be added to the above regimens 
The chemotherapy regimen chosen should attempt to balance the desire for disease reduction/control with the risk of infections or toxicities that could delay or preclude infusion.44,45 When possible, would be ideal to allow for count recovery in between bridging chemotherapy and CD19 CAR T-cell infusion. The addition of IT chemotherapy is crucial for CNS prophylaxis or to treat CNS involvement. 
Immunotherapy Blinatumomab; inotuzumab ozogamicin Blinatumomab: risk for CD19 antigen downregulation or escape, which may lead to worse outcomes after CAR T cells or even preclude the use of CD19 CAR T cells.18,20,26 Thus, we recommend against the immediate use of blinatumomab as a bridge to CD19 CAR T cells.
Inotuzumab: Can induce prolonged BCA. We recommend cautious use of inotuzumab immediately before CD19 CAR T cells, given data showing CD19+ antigen load may be associated with CAR T-cell persistence.32 In addition, for patients who are planning to proceed to consolidative HSCT after CD19 CAR T cells, inotuzumab is associated with increased risk of sinusoidal obstructive syndrome.46  
Radiotherapy No standard dosing protocols available May be considered for patients with bulky EMD not responsive to chemotherapy; minimal data available on its efficacy before CAR T-cell therapy.47,48  
Treatment modalityTherapiesConsiderations
Timing to apheresis  The following factors should be taken into consideration to determine optimal timing for apheresis:
Quantity of peripheral T cells: a minimum absolute lymphocyte count of >300 cells per μL or CD3+ cell count of 100 to 200 cells per μL is recommended.
Circulating peripheral blasts: it is generally not advisable to collect with a high circulating blast count (WBC > 20-40 000/μL with >50%-80% leukemic blasts) because this can interfere with manufacturing.42
Quantity of prior therapy exposure: we recommend collecting T cells as early as feasible because continued courses of chemotherapy selectively deplete early lineage cells, which are important for T-cell expansion.40,41 
Time from prior therapy: a minimum treatment-free period is needed before collection, ranging from 72 h for corticosteroids, to 2 wk for cytotoxic chemotherapy, to longer periods for certain immunotherapies (eg, 6 wk-3 mo for pembrolizumab).
Patients with a history of HSCT: we recommend that apheresis occur at least 100 to 120 d after HSCT and immunosuppression be discontinued at least 4 wk before apheresis without signs of graft-versus-host disease to avoid the risk of collecting alloreactive T-cells. 
Chemotherapy Intensive regimens: 4-drug induction (vincristine, corticosteroid, asparaginase and anthracycline); cyclophosphamide with etoposide; high-dose cytarabine.
Less-intensive regimens: maintenance-style chemotherapy (oral mercaptopurine, oral methotrexate, ±vincristine, and short course of corticosteroids); abbreviated consolidation cycle with cyclophosphamide and low-dose cytarabine; intermediate-dose VP-16/cytosine arabinoside; intermediate-dose methotrexate; 3-drug induction (vincristine, corticosteroid, and asparaginase)
IT chemotherapy should be added to the above regimens 
The chemotherapy regimen chosen should attempt to balance the desire for disease reduction/control with the risk of infections or toxicities that could delay or preclude infusion.44,45 When possible, would be ideal to allow for count recovery in between bridging chemotherapy and CD19 CAR T-cell infusion. The addition of IT chemotherapy is crucial for CNS prophylaxis or to treat CNS involvement. 
Immunotherapy Blinatumomab; inotuzumab ozogamicin Blinatumomab: risk for CD19 antigen downregulation or escape, which may lead to worse outcomes after CAR T cells or even preclude the use of CD19 CAR T cells.18,20,26 Thus, we recommend against the immediate use of blinatumomab as a bridge to CD19 CAR T cells.
Inotuzumab: Can induce prolonged BCA. We recommend cautious use of inotuzumab immediately before CD19 CAR T cells, given data showing CD19+ antigen load may be associated with CAR T-cell persistence.32 In addition, for patients who are planning to proceed to consolidative HSCT after CD19 CAR T cells, inotuzumab is associated with increased risk of sinusoidal obstructive syndrome.46  
Radiotherapy No standard dosing protocols available May be considered for patients with bulky EMD not responsive to chemotherapy; minimal data available on its efficacy before CAR T-cell therapy.47,48  

IT, intrathecal; WBC, white blood cell.

Bridging therapy

Once a patient undergoes apheresis, bridging therapy during the manufacturing period needs to be considered based on the extent of the underlying disease and the anticipated time to CAR T-cell infusion. The intensity of this therapy should be individualized to avoid toxicities that could delay or preclude CAR T-cell infusion. Recognizing the association of high disease burden with poor CAR T-cell outcomes,20,27,30 the desire to give more intensive therapies to reduce the preinfusion disease burden has to be balanced against the risk of infection, organ damage, or cytopenias, and the likelihood that intensification will not result in CR in highly chemorefractory disease. In addition, 1 study showed the importance of having at least 15% of bone marrow cells (normal B cells or blasts) expressing CD19+ before therapy to optimize CAR T-cell expansion and durability.32 Therefore, for patients in remission or with central nervous system (CNS)–only disease, minimal or no bridging therapy to allow for normal B-cell expansion in the peripheral blood is a reasonable approach. Various bridging therapies, including chemotherapy, radiation therapy, and immunotherapy, have been used before CAR T cells, and specific considerations are summarized in Table 2.44,45,47 

Case (continued): after successful apheresis, the patient receives maintenance-style chemotherapy during CAR T-cell manufacturing. Preinfusion marrow disease burden was 30% without evidence of CNS or testicular involvement.

Marrow disease burden

High bone marrow disease burden at the time of infusion is the most important risk factor associated with toxicity.49-51 In addition, high disease burden is also highly prognostic for worse long-term RFS.20,27-30 Unfortunately, there is not a standard definition for “high disease burden.” The initial analyses of cytokine release syndrome (CRS) after tisagenlecleucel identified 40% of bone marrow blasts as the threshold for severe CRS risk.50,52 Most analyses of CAR response identified 5% of bone marrow blasts as the threshold for increased relapse and worse RFS.20,27,29,30 The association of high disease burden with inferior outcomes is striking. In a report from the PRWCC (n = 185), patients with bone marrow blasts >5% had 12-month EFS and OS rates of 31% and 58%, compared with 70% and 85% for those with low disease burden, respectively (P < .0001).27 A report from the CAR-MA study (n = 420) further demonstrated that these poor outcomes (Figure 1B) were specifically driven by a higher cumulative incidence of CD19 relapse (HR 5.2, P < .001).23 These compelling findings may identify a subset for whom CAR T cells may not lead to long-term, durable remissions and where post–CAR T-cell consolidative HSCT could be considered. Although disease burden is a potentially modifiable variable, intensifying bridging therapy may be counterproductive and lead to more toxicity; a careful study of this issue would be worthwhile.

CNS disease burden

For CNS disease, because of concerns about immune effector cell–associated neurotoxicity syndrome (ICANS), most early CD19 CAR T-cell trials excluded patients with bulk CNS tumors or CNS3 disease. More recently, however, data have been published showing that patients with CNS disease who undergo CAR T-cell therapy have similar outcomes to those without CNS disease, with no increase in severe ICANS,31,53 but mixed results on the risk of relapse.54 Based on these data and our clinical experience, for patients with CNS disease at referral, we recommend: (1) treating patients with intrathecal and sometimes CNS-targeted systemic therapy to ensure stable/responding disease at the time of CAR T-cell infusion; (2) proceeding cautiously (following optimal preinfusion treatment for risk mitigation) in patients with persistent CNS parenchymal lesions, as this may increase ICANS risk based on emerging data from CNS tumors and CAR T cells55 and until more data in B-ALL is available; (3) allowing a 4- to 8-week interval from cranial radiation to infusion for patients requiring focal or more general cranial irradiation to provide distance from potential side effect of CNS radiation (eg, somnolence syndrome)56 that could mimic symptoms of ICANS; and (4) ensuring no acute or worsening neurologic toxicity at the time of infusion. The use of antiepileptics in patients with preexisting CNS disease or prior neurotoxicity (eg, methotrexate toxicity) should be considered to mitigate risk of seizures.57 

Non-CNS extramedullary disease (EMD)

For non-CNS EMD, the available data is limited. A large retrospective analysis found that active EMD during infusion was independently associated with worse EFS (HR 1.9, P = .01).20 A PRWCC analysis found that in 15 patients with non-CNS EMD, 10 achieved a CR, and 6 of the 10 subsequently relapsed. Consequently, only 4 of 15 patients remained event-free during the follow-up period.31 A retrospective analysis from the NCI identified limitations in the eradication of non-CNS EMD with CAR T cells, despite concurrent marrow responses, highlighting the important role of positron emission tomography scans for monitoring.58 Reducing bulky EMD with radiation or chemotherapy before infusion may improve overall outcomes, but residual disease should not preclude CAR T-cell treatment. Given little data regarding CAR T-cell trafficking to and efficacy with testicular disease, we recommend considering definitive treatment of testicular disease before infusion.

Lymphodepletion

Lymphodepletion is essential to provide an in vivo environment that will enhance the expansion, function, and persistence of CAR T cells, and most regimens include fludarabine and cyclophosphamide (Table 2), based on data from the pivotal CD19 CAR trials.47 Recent data suggests there may be room to further optimize these regimens. Two retrospective studies found that suboptimal fludarabine exposure, defined as a measured or estimated area under the curve <13.8 to 14.0 mg × h/L, was associated with increased relapse risk and a higher probability of premature loss of B-cell aplasia (BCA).33,34 Although there is insufficient data to change standard lymphodepletion regimens, optimizing lymphodepleting agents is an area where prospective, comparative trials may lead to improved outcomes, especially in light of recent drug shortages.59 

Case (continued): the patient received tisagenlecleucel and experienced grade 2 CRS without ICANS. On day +28, he was MRD by flow and had achieved BCA. Next-generation sequencing (NGS)-MRD was sent and is positive at <1 cell, below the level of quantitation of 1 × 10−6.

Once remission has been obtained after tisagenlecleucel, based upon long-term data from the ELIANA trial (N = 75), about half of patients will have continued remission without interval therapy and possibly be cured at 5 years; however, the other half will relapse. Given limited salvage options for CAR T-cell relapse23,60 particularly for LS (Figure 1C), clinicians are faced with a choice: send all patients empirically to HSCT, selectively move to HSCT for high-risk patients only, or reserve HSCT for patients who relapse and get back into remission. In the absence of studies designed to define best outcomes, we will review the first 2 of these options while sharing available data.

Consolidative HSCT for all patients receiving CD19 CAR T cells

For patients receiving CD19-directed CAR T-cell constructs, where long-term remissions are not expected (eg, CD28-based CAR T cells), HSCT is critical. Differing from conclusions of an early study of CD28 CAR T cells in adults where OS with or without HSCT was poor,61 consolidative HSCT usually leads to long-term, durable remission in younger patients.10 The challenge is deciding when to take patients to HSCT after treatments similar to tisagenlecleucel, which have curative potential. A recent meta-analysis addressed this issue,62 but the trials in the analysis included mainly adult patients, used constructs that had not demonstrated efficacy for long-term responses without HSCT, and planned for HSCT rather than randomly assign patients or try to define patients who could avoid HSCT. Two pediatric studies give better insight into this question. Investigators in Seattle looked retrospectively at their outcomes with consolidative HSCT after CAR T cells. In 50 patients potentially eligible for HSCT after achieving and maintaining an MRD remission beyond day 63 after a 4-1BB CAR T-cell construct with long-term curative potential, HSCT-naïve patients receiving a post–CAR consolidative HSCT had a trend toward improved leukemia-free survival (LFS) (P = .09), whereas those with a history of prior HSCT had no advantage in LFS (P = .45). No advantage was noted in OS, as many patients were salvaged with additional therapy. Patients who had loss of BCA/CAR T-cell disappearance by day 63 and received a consolidative first HSCT showed a clear improvement in LFS (P = .01).63 A study from China of children receiving either CD19- or CD22-directed CAR T-cell therapy showed a 1-year EFS of 73% with planned HSCT,64 which compares to approximately 50% EFS in the ELIANA study4 and other real-world27 or collaborative reports.20 These studies suggest an advantage for HSCT after CAR T cells only in HSCT-naïve patients, especially if they lose BCA early, but the experiences are small, single-center, and nonrandomized.

Consolidative HSCT for high-risk patients

Given the identification of risk factors affecting outcomes (Table 1) and poor salvage rates after relapse,23 we are now better informed about patients at high risk of poor outcomes. But there is a sizable portion of patients in poor risk groups who can be cured with tisagenlecleucel alone, so post–CAR T-cell risk factors to better predict patients who will fail are important. To date, there have been only 2 factors post–CAR T-cell therapy that reliably predict relapse: loss of BCA and detection of NGS-MRD.

BCA is a functional screen for active CAR T cells, and loss of BCA before 6 months is associated with very high rates of relapse.49 The measure is suboptimal, however, as patients who relapse with CD19-negative ALL can relapse despite CAR T-cell persistence.23 Because CD19-negative relapse occurs frequently, measuring for BCA alone is inadequate. In addition, the power of BCA to predict outcome fades with time, with 1 study showing that loss of BCA in the first 6 months led to an EFS rate lower than 20%, whereas patients who lost BCA at 1 year had an EFS rate of 75%.36 

NGS-MRD can potentially overcome the limitations of following BCA alone by tracking the leukemia genomic clone directly, detecting both CD19+ and CD19 disease. Using NGS of blast-specific immunoglobulin heavy chain (IgH) rearrangements to test for MRD after CAR T-cell therapy, considering any level of disease detected positive, multivariate analysis at day 28 showed an independent association of NGS-MRD with relapse (HR, 4.87; 95% confidence interval, 2.18-10.8; P < .001), which increased at 3 months to a HR of 12 (95% confidence interval, 2.87-50; P < .001; Figure 2).36 Although some may consider NGS-MRD detection of blasts simply a more sensitive measure of relapse, it is not part of current definitions of relapse.65 Detection at day 28 after CAR T-cell therapy can be part of an ongoing response and should be verified (Figure 2A). By 3 months, however, any presence of NGS-MRD is highly predictive of relapse (Figure 2C). Another study showed that detection of disease by IG/T-cell receptor RQ-polymerase chain reaction (PCR)–based MRD at day +28 after CAR T-cell therapy is also highly predictive22 and can be used in situations where NGS-MRD is not available.

Figure 2.

EFS and OS in patients in CR/CRi following tisagenlecleucel by Kaplan-Meier analyses with log-rank test P values. (A-B) EFS (A) and OS (B) of responding patients based on detection at 28 days after CAR infusion of BM NGS-MRD at any level (blue lines) compared with patients with BM NGS-MRD = 0 (green lines). (C-D) EFS (C) and OS (D) of responding patients based on detection at 3 months after CAR infusion of BM NGS-MRD at any level (blue lines) compared with patients with BM NGS-MRD = 0 (green lines). CRi, complete remission with incomplete count recovery. Reproduced with permission from CCC Marketplace, license number 1257504-1, from Pulsipher et al.36 

Figure 2.

EFS and OS in patients in CR/CRi following tisagenlecleucel by Kaplan-Meier analyses with log-rank test P values. (A-B) EFS (A) and OS (B) of responding patients based on detection at 28 days after CAR infusion of BM NGS-MRD at any level (blue lines) compared with patients with BM NGS-MRD = 0 (green lines). (C-D) EFS (C) and OS (D) of responding patients based on detection at 3 months after CAR infusion of BM NGS-MRD at any level (blue lines) compared with patients with BM NGS-MRD = 0 (green lines). CRi, complete remission with incomplete count recovery. Reproduced with permission from CCC Marketplace, license number 1257504-1, from Pulsipher et al.36 

Close modal

Unfortunately, when using NGS-MRD, about 10% of patients with B-ALL will not have informative IgH clones, as used in the aforementioned study. Use of T-cell receptor clones may improve that number to >95%, but these clones vary in specificity.66 Accessibility to commercial NGS-MRD testing is another substantial limitation. Consultation with experts in this assay may be needed for suspicious results. Key factors strengthening the predictive power of this test include positivity in multiple clones and a clear pattern of increase in clonal numbers with serial testing.

Given the need to prospectively study these critical biomarkers, the upcoming CAR-CURE trial (ClinicalTrials.gov NCT05621291) will test the hypothesis that systematic surveillance of BCA and NGS-MRD measurements after CD19 CAR T cells can identify those at highest risk for relapse after CAR T-cell infusion and route them to HSCT before relapse.67 

Approaches to patients treated with CAR T cells for relapse after HSCT

Given the risks and complications associated with second HSCT,68 post–CAR T-cell consolidative second HSCTs cannot be routinely recommended. Selecting a more persistent CAR T cell is recommended for these very high-risk patients, for whom curative options are limited. Data reporting on outcomes for second HSCT after CAR T-cell–induced remission are based on small cohorts and do not show the same degree of benefit of HSCT compared with transplant-naïve patients.69,70 Therefore, for these patients, experimental CAR T-cell approaches aimed at improving cure as a single therapy or strategies aimed at preventing relapse should be encouraged, with second HSCT reserved only for those clearly at imminent risk of relapse.

Case (continued): repeat IgH NGS-MRD remains low-level positive at day +60 with concurrent loss of BCA as the absolute CD19+ count rises to 75/μL. The HSCT team plans to move forward with the transplant as soon as possible, and a fully matched unrelated donor has been identified.

Bridging high-risk patients to HSCT

With early loss of BCA and/or persistent or rising NGS-MRD, relapse may be imminent.36 Because of the high rates of survival when patients are NGS-MRD before HSCT,71 initiating therapy to induce or maintain NGS-MRD negativity is often considered, particularly when awaiting donor availability. Given the chemotherapy refractory nature of the disease in patients who received CAR T cells and because NGS-MRD cannot elucidate the leukemic immunophenotype, bridging options are both limited and empiric. Blinatumomab may be particularly effective with low disease burden if CD19+ is retained,72,73 but as antigen expression is not known with NGS-MRD, efficacy may be limited if the disease is CD19. Inotuzumab ozogamicin, a CD22-targeted conjugated antibody, is a consideration but particularly problematic in the peri-HSCT setting, given concerns for veno-occlusive disease/sinusoidal obstructive syndrome.46,74 Nonimmunotherapeutic and antigen agnostic chemotherapy could also be used, though with caution given the general chemotherapy refractory nature of disease in patients receiving CAR T cells.

Proceeding to HSCT in patients who are RQ-PCR+ or NGS-MRD+ and flow MRD–negative may not be ideal, but it may be reasonable, as HSCT may be the only curative option and treatment of the residual disease may not be feasible or effective. When pre-HSCT flow cytometry is <0.01% or PCR is <10−4, survival is clearly improved.75 However, in patients who are NGS-MRD+ but flow cytometry–negative, developing some level of acute graft-versus-host disease and undergoing a myeloablative total body irradiation–based conditioning may be necessary to optimize chances of long-term survival.71 

CAR T-cell reinfusions for relapse prevention

The role of CAR T-cell reinfusion as a relapse prevention strategy for patients with early loss of BCA has not been firmly established, and patients losing CAR T cells before 6 months rarely have long-term responses. Several groups have reported limited success with reinfusion.32,76,77 However, investigators at Children’s Hospital of Philadelphia, using an approach of early preemptive reinfusions, have reported greater success.78 Among 63 patients reinfused for peripheral B-cell recovery (n = 38) or hematogones in the bone marrow (n = 25), 33 (52%) established or maintained BCA 28 days after reinfusion. Of those, 16 (48%) remained in remission without further therapy at a median of 39 months from reinfusion.78 The addition of checkpoint inhibitors after infusion has been suggested,79 but data showing a clear benefit is lacking.

Case (continued): because the identified unrelated donor had been confirmed, the patient started his preparative regimen for HSCT within 3 weeks with no interval therapy. He developed grade II acute graft-versus-host disease and no chronic graft-versus-host disease, and at 2 years post-HSCT, he remains in an ongoing remission.

Although CD19 CAR T cells have transformed the treatment of pediatric r/r B-ALL, applying risk factors associated with the success or failure of this therapy to optimize outcomes is a crucial next step. Although we use a single case to illustrate these important considerations, unique populations (eg, infants, those with early CNS relapse, etc) warrant unique considerations, which are outlined in Table 3. Given the potential for long-term cure with CD19 CAR T cells, avoiding allogeneic HSCT and its associated short- and long-term toxicities is greatly desired. However, given that treatment outcomes of relapse after CD19 CAR T cells are dismal,23 it is critical to define the best approaches to prevent relapse in patients at highest risk. In patients for whom consolidative HSCT following CAR T cells is not an option (eg, patient/parent preference or HSCT-limiting toxicities), developing better relapse-prevention strategies is critical. Treatment of pediatric B-ALL has always been built upon risk stratification, and based on current data, we have included our personalized approach to risk stratification of this therapy (Figure 3). We recommend prospective studies to validate and refine approaches aimed at improving outcomes following CD19 CAR T cells in pediatric B-ALL.

Table 3.

Special populations and unique patient scenarios for CD19 CAR T-cell therapy

Special population or unique scenarioSummary and recommendationsAvailable data
Isolated CNS relapse of B-ALL CAR T cells are effective in eradication of CNS disease in B-ALL.
Consider CAR T cells in:
Multiply relapsed isolated CNS disease.
Early isolated CNS relapse (not currently an approved indication for tisagenlecleucel)
Given favorable outcomes with standard of care therapy for late, isolated first relapse,80 CAR T cells would not be considered first line in this setting. 
Several retrospective studies described outcomes after CD19 CAR T cells for B-ALL with CNS involvement using aggregate data sets. Importantly, severe neurotoxicity was not found to be increased in patients with CNS involvement.31,53,54,
CHOP clinical trials: in a pooled analysis of 5 clinical trials at CHOP, the CR rate for patients treated with murine or humanized CD19 CAR T cells for isolated CNS disease (n = 43) was 98%.53 Of those, 38% experienced a subsequent relapse (7/16 with CNS involvement at post-CAR relapse); 2-y OS was 91%.
PRWCC study: the CR rate for patients treated with tisagenlecleucel for isolated CNS disease (n = 23) was 96%.31 Of those, 32% experienced a subsequent relapse (4/7 with CNS involvement at post-CAR relapse); 2-y OS was 81%.
International experience: in a multicenter study of 55 patients with CNS-positive disease treated with a variety of CD28 or 4-1BB CD19 CAR T cells, the CR rate was 94%.54 The 2-y EFS and OS were 40% and 75% for the full cohort, and 44% and 93% for those with isolated CNS disease (n = 15). Notably, among 8 patients who received tisagenlecleucel for isolated CNS disease, 6 subsequently relapsed (all in the CNS), but 2-y OS was 100%. 
Primary refractory disease High-risk patients with primary refractory disease should be considered for early referral to CAR T cells.
The role for consolidative HSCT for patients who receive CAR T cells for primary refractory disease warrants prospective study. 
As most clinical trials have enrolled patients with both r/r disease, isolating outcomes for those with primary refractory disease is challenging. Nonetheless, in patients with primary refractory disease across either early phase studies20 or pivotal trials,6 substantial benefit has been seen.
An ongoing, multicenter, phase 2 clinical trial through the Children’s Oncology Group (NCT03876769) is evaluating long-term outcomes following CD19 CAR T cells in high-risk patients with detectable MRD by flow cytometry after 2 cycles of frontline chemotherapy.
Given excellent outcomes with chemotherapy alone81 patients with NCI standard-risk B-ALL who are MRD+ after 2 cycles of chemotherapy, are presently excluded from the aforementioned study.
The role of consolidative HSCT is ill defined; the COG study is testing use of CD19 CAR T cells in an earlier setting when consolidative HSCT may or may not be needed. 
First relapse of ALL Clinical trials evaluating the role for CD19 CAR T cells are urgently needed given the limited remission rates and toxicity associated with chemotherapy-based induction regimens for patients with first relapse. Tisagenlecleucel is not currently approved for patients with first relapse. 
History of previous HSCT CAR T cells are an effective salvage treatment for patients with post-HSCT relapse of B-ALL.
Given the risks associated with second HSCT,68 CAR T cells with curative potential should be preferentially considered to try to avoid the need for a consolidative second HSCT. 
There are limited effective options for patients with post-HSCT relapsed B-ALL. A large proportion of patients treated with CAR T cells have had a history of prior HSCT.
Multiple studies have shown that overall outcomes are similar among patients with or without a history of prior HSCT.20,27  
DS CAR T cells are a reasonable consideration for patients with relapsed DS-associated ALL, especially in light of inferior outcomes with standard-of-care therapies.82  Individual CAR T-cell trials have reported small numbers of patients with DS-associated ALL.
Pooled ELIANA/ENSIGN analysis: in a post hoc analysis of 16 patients with DS treated on 2 phase 2 trials and a phase 3b managed access protocol, 14 (88%) achieved a CR and 9 of 14 (64%) remained in remission with a median follow-up of 13.2 mo.37 There was no increased toxicity risk compared with patients without DS. 
Infant B-ALL CAR T cells are a reasonable consideration for patients with relapsed, infant B-ALL, especially in light of poor outcomes with standard-of-care therapies.
Despite comparable relapse risks to other patients receiving CAR T cells, the risk of lineage switch is markedly higher with no long-term survivors reported after lineage switch.
The role of post–CAR T-cell consolidative HSCT for these patients warrants further study as we do not know if it can prevent lineage switch. Further, avoidance of toxicities associated with total body radiation in young children make it challenging to deliver an optimal HSCT. 
Several retrospective studies describe outcomes after CAR T cells for infant, KMT2A-rearranged B-ALL.
CHOP clinical trials: in an analysis of 13 patients from CHOP, RFS was comparable to other patients with a 2-y RFS of 67% (HR 0.70; P = .704), but OS was worse (HR 3.6, P = .043) because of limited ability to salvage after post-CAR relapse.38 Notably, 3 patients experienced myeloid lineage switch and none survived.

CAR-MA: of 29 patients with infant B-ALL, 25 (86%) achieved a CR, but 9 of 29 (31%) had a subsequent relapse; 6 relapses included myeloid lineage switch and none of those patients survived.23 In a PRWCC analysis, of 14 patients with infant B-ALL, 9 achieved an MRD CR (64%), of which 7 remain in remission at a median follow-up of 290 d. Lineage switch was observed in 4 patients, 3 of which had a nonresponse to CAR T cells.39,
European experience: 35 patients <3 y old at the time of screening for tisagenlecleucel, 31 (89%) achieved an MRD CR, of which 21 (68%) remain in remission at a median follow-up of 14 mo.83 Lineage switch was not observed. 
ALL with targetable mutations Patients with BCR-ABL or other targetable mutations may benefit from use of these agents for disease control before CAR T-cell therapy.
Careful consideration must be given to the effect of targeted therapy on collected apheresis products.
Most targeted therapies have a clear anti–T-cell effect and should be avoided after CAR T-cell therapy, unless the patient has had loss of BCA. 
 
Special population or unique scenarioSummary and recommendationsAvailable data
Isolated CNS relapse of B-ALL CAR T cells are effective in eradication of CNS disease in B-ALL.
Consider CAR T cells in:
Multiply relapsed isolated CNS disease.
Early isolated CNS relapse (not currently an approved indication for tisagenlecleucel)
Given favorable outcomes with standard of care therapy for late, isolated first relapse,80 CAR T cells would not be considered first line in this setting. 
Several retrospective studies described outcomes after CD19 CAR T cells for B-ALL with CNS involvement using aggregate data sets. Importantly, severe neurotoxicity was not found to be increased in patients with CNS involvement.31,53,54,
CHOP clinical trials: in a pooled analysis of 5 clinical trials at CHOP, the CR rate for patients treated with murine or humanized CD19 CAR T cells for isolated CNS disease (n = 43) was 98%.53 Of those, 38% experienced a subsequent relapse (7/16 with CNS involvement at post-CAR relapse); 2-y OS was 91%.
PRWCC study: the CR rate for patients treated with tisagenlecleucel for isolated CNS disease (n = 23) was 96%.31 Of those, 32% experienced a subsequent relapse (4/7 with CNS involvement at post-CAR relapse); 2-y OS was 81%.
International experience: in a multicenter study of 55 patients with CNS-positive disease treated with a variety of CD28 or 4-1BB CD19 CAR T cells, the CR rate was 94%.54 The 2-y EFS and OS were 40% and 75% for the full cohort, and 44% and 93% for those with isolated CNS disease (n = 15). Notably, among 8 patients who received tisagenlecleucel for isolated CNS disease, 6 subsequently relapsed (all in the CNS), but 2-y OS was 100%. 
Primary refractory disease High-risk patients with primary refractory disease should be considered for early referral to CAR T cells.
The role for consolidative HSCT for patients who receive CAR T cells for primary refractory disease warrants prospective study. 
As most clinical trials have enrolled patients with both r/r disease, isolating outcomes for those with primary refractory disease is challenging. Nonetheless, in patients with primary refractory disease across either early phase studies20 or pivotal trials,6 substantial benefit has been seen.
An ongoing, multicenter, phase 2 clinical trial through the Children’s Oncology Group (NCT03876769) is evaluating long-term outcomes following CD19 CAR T cells in high-risk patients with detectable MRD by flow cytometry after 2 cycles of frontline chemotherapy.
Given excellent outcomes with chemotherapy alone81 patients with NCI standard-risk B-ALL who are MRD+ after 2 cycles of chemotherapy, are presently excluded from the aforementioned study.
The role of consolidative HSCT is ill defined; the COG study is testing use of CD19 CAR T cells in an earlier setting when consolidative HSCT may or may not be needed. 
First relapse of ALL Clinical trials evaluating the role for CD19 CAR T cells are urgently needed given the limited remission rates and toxicity associated with chemotherapy-based induction regimens for patients with first relapse. Tisagenlecleucel is not currently approved for patients with first relapse. 
History of previous HSCT CAR T cells are an effective salvage treatment for patients with post-HSCT relapse of B-ALL.
Given the risks associated with second HSCT,68 CAR T cells with curative potential should be preferentially considered to try to avoid the need for a consolidative second HSCT. 
There are limited effective options for patients with post-HSCT relapsed B-ALL. A large proportion of patients treated with CAR T cells have had a history of prior HSCT.
Multiple studies have shown that overall outcomes are similar among patients with or without a history of prior HSCT.20,27  
DS CAR T cells are a reasonable consideration for patients with relapsed DS-associated ALL, especially in light of inferior outcomes with standard-of-care therapies.82  Individual CAR T-cell trials have reported small numbers of patients with DS-associated ALL.
Pooled ELIANA/ENSIGN analysis: in a post hoc analysis of 16 patients with DS treated on 2 phase 2 trials and a phase 3b managed access protocol, 14 (88%) achieved a CR and 9 of 14 (64%) remained in remission with a median follow-up of 13.2 mo.37 There was no increased toxicity risk compared with patients without DS. 
Infant B-ALL CAR T cells are a reasonable consideration for patients with relapsed, infant B-ALL, especially in light of poor outcomes with standard-of-care therapies.
Despite comparable relapse risks to other patients receiving CAR T cells, the risk of lineage switch is markedly higher with no long-term survivors reported after lineage switch.
The role of post–CAR T-cell consolidative HSCT for these patients warrants further study as we do not know if it can prevent lineage switch. Further, avoidance of toxicities associated with total body radiation in young children make it challenging to deliver an optimal HSCT. 
Several retrospective studies describe outcomes after CAR T cells for infant, KMT2A-rearranged B-ALL.
CHOP clinical trials: in an analysis of 13 patients from CHOP, RFS was comparable to other patients with a 2-y RFS of 67% (HR 0.70; P = .704), but OS was worse (HR 3.6, P = .043) because of limited ability to salvage after post-CAR relapse.38 Notably, 3 patients experienced myeloid lineage switch and none survived.

CAR-MA: of 29 patients with infant B-ALL, 25 (86%) achieved a CR, but 9 of 29 (31%) had a subsequent relapse; 6 relapses included myeloid lineage switch and none of those patients survived.23 In a PRWCC analysis, of 14 patients with infant B-ALL, 9 achieved an MRD CR (64%), of which 7 remain in remission at a median follow-up of 290 d. Lineage switch was observed in 4 patients, 3 of which had a nonresponse to CAR T cells.39,
European experience: 35 patients <3 y old at the time of screening for tisagenlecleucel, 31 (89%) achieved an MRD CR, of which 21 (68%) remain in remission at a median follow-up of 14 mo.83 Lineage switch was not observed. 
ALL with targetable mutations Patients with BCR-ABL or other targetable mutations may benefit from use of these agents for disease control before CAR T-cell therapy.
Careful consideration must be given to the effect of targeted therapy on collected apheresis products.
Most targeted therapies have a clear anti–T-cell effect and should be avoided after CAR T-cell therapy, unless the patient has had loss of BCA. 
 

CHOP, Children’s Hospital of Philadelphia; DS, Down syndrome.

Figure 3.

Approach to peri-CAR T-cell risk stratification with monitoring and treatment options. This figure highlights various preinfusion risk factors that may affect long-term EFS, remission durability, and/or OS; and how these factors are further modulated by post–CAR T-cell outcomes, based primarily on NGS-MRD status and loss of BCA. Refer to Table 1 for specific information on risk factors. ASAP, as soon as possible; Blina, blinatumomab; CRi, complete remission with incomplete count recovery.

Figure 3.

Approach to peri-CAR T-cell risk stratification with monitoring and treatment options. This figure highlights various preinfusion risk factors that may affect long-term EFS, remission durability, and/or OS; and how these factors are further modulated by post–CAR T-cell outcomes, based primarily on NGS-MRD status and loss of BCA. Refer to Table 1 for specific information on risk factors. ASAP, as soon as possible; Blina, blinatumomab; CRi, complete remission with incomplete count recovery.

Close modal

Figure 3 was created with Biorender.com. The authors acknowledge Elizabeth Holland (National Institutes of Health [NIH]) for her support in developing Figure 3.

R.M.M. is supported by NIH, National Cancer Institute (NCI) grant K-12-CA-076931 and the Alex’s Lemonade Stand Foundation. M.A.P. is supported by NIH, National Institute of Allergy and Infectious Diseases grant 1U01AI126612-01A1, NIH, NCI grant P30CA040214, and the St. Baldrick’s Johnny Chrisstopher Fund. This work was supported in part by the Intramural Research Program, Center of Cancer Research, NCI and NIH Clinical Center, and National Institutes of Health (ZIA BC 011823, N.N.S.).

The content of this publication does not necessarily reflect the views of policies of the Department of Health and Human Services, nor does mention of trade names, commercial products, or organizations imply endorsement by the US government.

Contribution: M.A.P. and N.N.S. conceived the proposed idea; and all authors contributed to outlining, writing, and editing the manuscript and approved the final version of the manuscript.

Conflict-of-interest disclosure: M.A.P. has participated in advisory boards for Novartis, Gentibio, Bluebird, Vertex, Medexus, Mesoblast, and Equillium. He has given educational talks for Novartis and Adaptive. He receives study support from Adaptive and Miltenyi. N.N.S. received royalties from Syncopation Life Sciences and has participated in advisory boards for Sobi and VOR. R.M.M. declares no competing financial interests.

Correspondence: Michael A. Pulsipher, Huntsman Cancer Institute, 2000 Circle of Hope Dr Rm 3515, Salt Lake City, UT 84112; e-mail: michael.pulsipher@hci.utah.edu.

1.
Grupp
SA
,
Kalos
M
,
Barrett
D
, et al
.
Chimeric antigen receptor-modified T cells for acute lymphoid leukemia
.
N Engl J Med
.
2013
;
368
(
16
):
1509
-
1518
.
2.
CHOP News. Emily Whitehead, first pediatric patient to receive CAR T-cell therapy, celebrates cure 10 years later; 11 May 2022
. https://www.chop.edu/news/emily-whitehead-first-pediatric-patient-receive-car-t-cell-therapy-celebrates-cure-10-years.
3.
O'Leary
MC
,
Lu
X
,
Huang
Y
, et al
.
FDA approval summary: tisagenlecleucel for treatment of patients with relapsed or refractory B-cell precursor acute lymphoblastic leukemia
.
Clin Cancer Res
.
2019
;
25
(
4
):
1142
-
1146
.
4.
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
(
5
):
439
-
448
.
5.
Bouchkouj
N
,
Lin
X
,
Wang
X
, et al
.
FDA Approval Summary: Brexucabtagene Autoleucel for Treatment of Adults With Relapsed or Refractory B-Cell Precursor Acute Lymphoblastic Leukemia
.
Oncologist
.
2022
;
27
(
10
):
892
-
899
.
6.
Shah
BD
,
Ghobadi
A
,
Oluwole
OO
, 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
(
10299
):
491
-
502
.
7.
Wayne
AS
,
Huynh V
,
Hijiya
N
, et al
. Phase 1 results of ZUMA-4: KTE-X19, an anti-CD19 chimeric antigen receptor T-cell therapy in pediatric and adolescent patients with relapsed/refractory B-cell acute lymphoblastic leukemia. EHA open access library - Abstract PS962.
European Hematology Association
;
2019
.
8.
Rives
S
,
Maude
SL
,
Hiramatsu
H
, 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
.
9.
Lee
DW
,
Kochenderfer
JN
,
Stetler-Stevenson
M
, 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
(
9967
):
517
-
528
.
10.
Shah
NN
,
Lee
DW
,
Yates
B
, 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
(
15
):
1650
-
1659
.
13.
Shah
BD
,
Bishop
MR
,
Oluwole
OO
, et al
.
KTE-X19 anti-CD19 CAR T-cell therapy in adult relapsed/refractory acute lymphoblastic leukemia: ZUMA-3 phase 1 results
.
Blood
.
2021
;
138
(
1
):
11
-
22
.
14.
Brown
PA
,
Ji
L
,
Xu
X
, 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
(
9
):
833
-
842
.
15.
Jen
EY
,
Xu
Q
,
Schetter
A
, et al
.
FDA approval: blinatumomab for patients with B-cell precursor acute lymphoblastic leukemia in morphologic remission with minimal residual disease
.
Clin Cancer Res
.
2019
;
25
(
2
):
473
-
477
.
16.
Pulte
ED
,
Vallejo
J
,
Przepiorka
D
, et al
.
FDA supplemental approval: blinatumomab for treatment of relapsed and refractory precursor B-cell acute lymphoblastic leukemia
.
Oncologist
.
2018
;
23
(
11
):
1366
-
1371
.
17.
Majzner
RG
,
Mackall
CL
.
Tumor antigen escape from CAR T-cell therapy
.
Cancer Discov
.
2018
;
8
(
10
):
1219
-
1226
.
18.
Mejstríková
E
,
Hrusak
O
,
Borowitz
MJ
, et al
.
CD19-negative relapse of pediatric B-cell precursor acute lymphoblastic leukemia following blinatumomab treatment
.
Blood Cancer J
.
2017
;
7
(
12
):
659
.
19.
Demosthenous
C
,
Lalayanni
C
,
Iskas
M
,
Douka
V
,
Pastelli
N
,
Anagnostopoulos
A
.
Extramedullary relapse and discordant CD19 expression between bone marrow and extramedullary sites in relapsed acute lymphoblastic leukemia after blinatumomab treatment
.
Curr Probl Cancer
.
2019
;
43
(
3
):
222
-
227
.
20.
Myers
RM
,
Taraseviciute
A
,
Steinberg
SM
, et al
.
Blinatumomab nonresponse and high-disease burden are associated with inferior outcomes after CD19-CAR for B-ALL
.
J Clin Oncol
.
2022
;
40
(
9
):
932
-
944
.
21.
Barz Leahy
A
,
Devine
KJ
,
Li
Y
, et al
.
Impact of high-risk cytogenetics on outcomes for children and young adults receiving CD19-directed CAR T cell therapy
.
Blood
.
2022;139(14):2173–2185
.
22.
Dourthe
ME
,
Rabian
F
,
Yakouben
K
, et al
.
Determinants of CD19-positive vs CD19-negative relapse after tisagenlecleucel for B-cell acute lymphoblastic leukemia
.
Leukemia
.
2021
;
35
(
12
):
3383
-
3393
.
23.
Lamble
A
,
Myers
RM
,
Taraseviciute
A
, et al
.
Preinfusion factors impacting relapse immunophenotype following CD19 CAR T cells
.
Blood Adv
.
2023
;
7
(
4
):
575
-
585
.
24.
Zhang
X
,
Lu
XA
,
Yang
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
(
10
):
2325
-
2338
.
25.
Weber
EW
,
Lynn
RC
,
Sotillo
E
,
Lattin
J
,
Xu
P
,
Mackall
CL
.
Pharmacologic control of CAR-T cell function using dasatinib
.
Blood Adv
.
2019
;
3
(
5
):
711
-
717
.
26.
Pillai
V
,
Muralidharan
K
,
Meng
W
, et al
.
CAR T-cell therapy is effective for CD19-dim B-lymphoblastic leukemia but is impacted by prior blinatumomab therapy
.
Blood Adv
.
2019
;
3
(
22
):
3539
-
3549
.
27.
Schultz
LM
,
Baggott
C
,
Prabhu
S
, 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
.
2022
;
40
(
9
):
945
-
955
.
28.
Kadauke
S
,
Myers
RM
,
Li
Y
, et al
.
Risk-adapted preemptive tocilizumab to prevent severe cytokine release syndrome after CTL019 for pediatric B-cell acute lymphoblastic leukemia: a prospective clinical trial
.
J Clin Oncol
.
2021
;
39
(
8
):
920
-
930
.
29.
Myers
RM
,
Li
Y
,
Barz Leahy
A
, et al
.
Humanized CD19-targeted chimeric antigen receptor (CAR) T cells in CAR-naive and CAR-exposed children and young adults with relapsed or refractory acute lymphoblastic leukemia
.
J Clin Oncol
.
2021
;
39
(
27
):
3044
-
3055
.
30.
Ravich
JW
,
Huang
S
,
Zhou
Y
, et al
.
Impact of high disease burden on survival in pediatric patients with B-ALL treated with tisagenlecleucel
.
Transplant Cell Ther
.
2022
;
28
(
2
):
73.e71
-
73.e79
.
31.
Fabrizio
VA
,
Phillips
CL
,
Lane
A
, et al
.
Tisagenlecleucel outcomes in relapsed/refractory extramedullary ALL: a pediatric real world CAR consortium report
.
Blood Adv
.
2022
;
6
(
2
):
600
-
610
.
32.
Gardner
RA
,
Finney
O
,
Annesley
C
, et al
.
Intent-to-treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults
.
Blood
.
2017
;
129
(
25
):
3322
-
3331
.
33.
Fabrizio
VA
,
Boelens
JJ
,
Mauguen
A
, et al
.
Optimal fludarabine lymphodepletion is associated with improved outcomes after CAR T-cell therapy
.
Blood Adv
.
2022
;
6
(
7
):
1961
-
1968
.
34.
Dekker
L
,
Calkoen
FG
,
Jiang
Y
, et al
.
Fludarabine exposure predicts outcome after CD19 CAR T-cell therapy in children and young adults with acute leukemia
.
Blood Adv
.
2022
;
6
(
7
):
1969
-
1976
.
35.
Ghilardi
G
,
Chong
EA
,
Svoboda
J
, et al
.
Bendamustine is safe and effective for lymphodepletion before tisagenlecleucel in patients with refractory or relapsed large B-cell lymphomas
.
Ann Oncol
.
2022
;
33
(
9
):
916
-
928
.
36.
Pulsipher
MA
,
Han
X
,
Maude
SL
, et al
.
Next-generation sequencing of minimal residual disease for predicting relapse after Tisagenlecleucel in children and young adults with acute lymphoblastic leukemia
.
Blood Cancer Discov
.
2022
;
3
(
1
):
66
-
81
.
37.
Laetsch
TW
,
Maude
SL
,
Balduzzi
A
, et al
.
Tisagenlecleucel in pediatric and young adult patients with Down syndrome-associated relapsed/refractory acute lymphoblastic leukemia
.
Leukemia
.
2022
;
36
(
6
):
1508
-
1515
.
38.
Leahy
AB
,
Devine
KJ
,
Li
Y
, et al
.
Impact of high-risk cytogenetics on outcomes for children and young adults receiving CD19-directed CAR T-cell therapy
.
Blood
.
2022
;
139
(
14
):
2173
-
2185
.
39.
Moskop
A
,
Pommert
L
,
Baggott
C
, et al
.
Real-world use of Tisagenlecleucel in infant acute lymphoblastic leukemia
.
Blood Adv
.
2022
;
6
(
14
):
4251
-
4255
.
40.
Das
RK
,
Vernau
L
,
Grupp
SA
,
Barrett
DM
.
Naive T-cell deficits at diagnosis and after chemotherapy impair cell therapy potential in pediatric cancers
.
Cancer Discov
.
2019
;
9
(
4
):
492
-
499
.
41.
Singh
N
,
Perazzelli
J
,
Grupp
SA
,
Barrett
DM
.
Early memory phenotypes drive T cell proliferation in patients with pediatric malignancies
.
Sci Transl Med
.
2016
;
8
(
320
):
320ra323
.
42.
Allen
ES
,
Stroncek
DF
,
Ren
J
, et al
.
Autologous lymphapheresis for the production of chimeric antigen receptor T cells
.
Transfusion
.
2017
;
57
(
5
):
1133
-
1141
.
43.
Shah
NN
,
Highfill
SL
,
Shalabi
H
, et al
.
CD4/CD8 T-cell selection affects chimeric antigen receptor (CAR) T-cell potency and toxicity: updated results from a phase I anti-CD22 CAR T-cell trial
.
J Clin Oncol
.
2020
;
38
(
17
):
1938
-
1950
.
44.
Perica
K
,
Flynn
J
,
Curran
KJ
, et al
.
Impact of bridging chemotherapy on clinical outcome of CD19 CAR T therapy in adult acute lymphoblastic leukemia
.
Leukemia
.
2021
;
35
(
11
):
3268
-
3271
.
45.
Shahid
S
,
Ramaswamy
K
,
Flynn
J
, et al
.
Impact of bridging chemotherapy on clinical outcomes of CD19-specific CAR T cell therapy in children/young adults with relapsed/refractory B cell acute lymphoblastic leukemia
.
Transplant Cell Ther
.
2022
;
28
(
2
):
72.e71
-
72.e78
.
46.
O'Brien
MM
,
Ji
L
,
Shah
NN
, 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
.
2022
;
40
(
9
):
956
-
967
.
47.
Amini
L
,
Silbert
SK
,
Maude
SL
, et al
.
Preparing for CAR T cell therapy: patient selection, bridging therapies and lymphodepletion
.
Nat Rev Clin Oncol
.
2022
;
19
(
5
):
342
-
355
.
48.
Marquez
CP
,
Violari
EG
,
Sodji
Q
, et al
.
Multidisciplinary management of newly diagnosed pediatric large cell neuroendocrine carcinoma of the lung causing hemoptysis
.
Pediatr Blood Cancer
.
2021
;
68
(
10
):
e29182
.
49.
Maude
SL
,
Frey
N
,
Shaw
PA
, et al
.
Chimeric antigen receptor T cells for sustained remissions in leukemia
.
N Engl J Med
.
2014
;
371
(
16
):
1507
-
1517
.
50.
Teachey
DT
,
Lacey
SF
,
Shaw
PA
, et al
.
Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia
.
Cancer Discov
.
2016
;
6
(
6
):
664
-
679
.
51.
Gofshteyn
JS
,
Shaw
PA
,
Teachey
DT
, et al
.
Neurotoxicity after CTL019 in a pediatric and young adult cohort
.
Ann Neurol
.
2018
;
84
(
4
):
537
-
546
.
52.
Maude
SL
,
Teachey
DT
,
Rheingold
SR
, et al
.
Sustained remissions with CD19-specific chimeric antigen receptor (CAR)-modified T cells in children with relapsed/refractory ALL
.
J Clin Oncol
.
2016
;
34
(
15 suppl
):
3011
.
53.
Leahy
AB
,
Newman
H
,
Li
Y
, et al
.
CD19-targeted chimeric antigen receptor T-cell therapy for CNS relapsed or refractory acute lymphocytic leukaemia: a post-hoc analysis of pooled data from five clinical trials
.
Lancet Haematol
.
2021
;
8
(
10
):
e711
-
e722
.
54.
Jacoby
E
,
Ghorashian
S
,
Vormoor
B
, et al
.
CD19 CAR T-cells for pediatric relapsed acute lymphoblastic leukemia with active CNS involvement: a retrospective international study
.
Leukemia
.
2022
;
36
(
6
):
1525
-
1532
.
55.
Majzner
RG
,
Ramakrishna
S
,
Yeom
KW
, et al
.
GD2-CAR T cell therapy for H3K27M-mutated diffuse midline gliomas
.
Nature
.
2022
;
603
(
7903
):
934
-
941
.
56.
Uzal
D
,
Ozyar
E
,
Hayran
M
,
Zorlu
F
,
Atahan
L
,
Yetkin
S
.
Reduced incidence of the somnolence syndrome after prophylactic cranial irradiation in children with acute lymphoblastic leukemia
.
Radiother Oncol
.
1998
;
48
(
1
):
29
-
32
.
57.
Maus
MV
,
Alexander
S
,
Bishop
MR
, et al
.
Society for immunotherapy of cancer (SITC) clinical practice guideline on immune effector cell-related adverse events
.
J Immunother Cancer
.
2020
;
8
(
2
):
e001511
.
58.
Holland
EM
,
Yates
B
,
Ling
A
, et al
.
Characterization of extramedullary disease in B-ALL and response to CAR T-cell therapy
.
Blood Adv
.
2022
;
6
(
7
):
2167
-
2182
.
59.
Maziarz
RT
,
Diaz
A
,
Miklos
DB
,
Shah
NN
.
Perspective: an international fludarabine shortage: supply chain issues impacting transplantation and immune effector cell therapy delivery
.
Transplant Cell Ther
.
2022
;
28
(
11
):
723
-
726
.
60.
Schultz
LM
,
Eaton
A
,
Baggott
C
, et al
.
Outcomes after nonresponse and relapse post-Tisagenlecleucel in children, adolescents, and young adults with B-cell acute lymphoblastic leukemia
.
J Clin Oncol
.
2023
;
41
(
2
):
354
-
363
.
61.
Park
JH
,
Riviere
I
,
Gonen
M
, et al
.
Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia
.
N Engl J Med
.
2018
;
378
(
5
):
449
-
459
.
62.
Xu
X
,
Chen
S
,
Zhao
Z
, et al
.
Consolidative hematopoietic stem cell transplantation after CD19 CAR-T cell therapy for acute lymphoblastic leukemia: a systematic review and meta-analysis
.
Front Oncol
.
2021
;
11
:
651944
.
63.
Summers
C
,
Wu
QV
,
Annesley
C
, et al
.
Hematopoietic cell transplantation after CD19 CAR T cell-induced ALL remission confers leukemia-free survival advantage
.
Transplant Cell Ther
.
2022;28(1):21-29
.
64.
Zhang
Y
,
Chen
H
,
Song
Y
, et al
.
Chimeric antigens receptor T cell therapy as a bridge to haematopoietic stem cell transplantation for refractory/relapsed B-cell acute lymphomalastic leukemia
.
Br J Haematol
.
2020
;
189
(
1
):
146
-
152
.
65.
Buchmann
S
,
Schrappe
M
,
Baruchel
A
, et al
.
Remission, treatment failure, and relapse in pediatric ALL: an international consensus of the Ponte-di-Legno consortium
.
Blood
.
2022
;
139
(
12
):
1785
-
1793
.
66.
Wood
B
,
Wu
D
,
Crossley
B
, et al
.
Measurable residual disease detection by high-throughput sequencing improves risk stratification for pediatric B-ALL
.
Blood
.
2018
;
131
(
12
):
1350
-
1359
.
67.
Heslop
HE
,
Stadtmauer
EA
,
Levine
JE
, et al
.
Blood and marrow transplant clinical trials network state of the science symposium 2021: looking forward as the network celebrates its 20th year
.
Transplant Cell Ther
.
2021
;
27
(
11
):
885
-
907
.
68.
Lund
TC
,
Ahn
KW
,
Tecca
HR
, et al
.
Outcomes after second hematopoietic cell transplantation in children and young adults with relapsed acute leukemia
.
Biol Blood Marrow Transplant
.
2019
;
25
(
2
):
301
-
306
.
69.
Molina
JC
,
Steinberg
SM
,
Yates
B
, et al
.
Factors impacting overall and event-free survival following post-chimeric antigen receptor T cell consolidative hematopoietic stem cell transplantation
.
Transplant Cell Ther
.
2022
;
28
(
1
):
31 e31
-
31 e39
.
70.
Summers
C
,
Wu
QV
,
Annesley
C
, et al
.
Hematopoietic cell transplantation after CD19 chimeric antigen receptor T cell-induced acute lymphoblastic lymphoma remission confers a leukemia-free survival advantage
.
Transplant Cell Ther
.
2022
;
28
(
1
):
21
-
29
.
71.
Pulsipher
MA
,
Carlson
C
,
Langholz
B
, et al
.
IgH-V(D)J NGS-MRD measurement pre- and early post-allotransplant defines very low- and very high-risk ALL patients
.
Blood
.
2015
;
125
(
22
):
3501
-
3508
.
72.
Jabbour
EJ
,
Short
NJ
,
Jain
N
, et al
.
Blinatumomab is associated with favorable outcomes in patients with B-cell lineage acute lymphoblastic leukemia and positive measurable residual disease at a threshold of 10(-4) and higher
.
Am J Hematol
.
2022;97(9):1135-1141
.
73.
Keating
AK
,
Gossai
N
,
Phillips
CL
, et al
.
Reducing minimal residual disease with blinatumomab prior to HCT for pediatric patients with acute lymphoblastic leukemia
.
Blood Adv
.
2019
;
3
(
13
):
1926
-
1929
.
74.
Pennesi
E
,
Michels
N
,
Brivio
E
, et al
.
Inotuzumab ozogamicin as single agent in pediatric patients with relapsed and refractory acute lymphoblastic leukemia: results from a phase II trial
.
Leukemia
.
2022
;
36
(
6
):
1516
-
1524
.
75.
Bader
P
,
Salzmann-Manrique
E
,
Balduzzi
A
, et al
.
More precisely defining risk peri-HCT in pediatric ALL: pre- vs post-MRD measures, serial positivity, and risk modeling
.
Blood Adv
.
2019
;
3
(
21
):
3393
-
3405
.
76.
Turtle
CJ
,
Hanafi
LA
,
Berger
C
, et al
.
CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients
.
J Clin Invest
.
2016
;
126
(
6
):
2123
-
2138
.
77.
Truscott
J
,
Lamble
A
,
Hsieh
E
, et al
.
Outcomes following CD19 CAR-T reinfusion in children and young adults with B-ALL. Abstracts from the 54th Congress of the International Society of Paediatric Oncology (SIOP) September 28 - October 1, 2022
.
Pediatr Blood Cancer
.
2022
;
69
(
Suppl 5
):
e29952
.
78.
Myers
RM
,
Devine
K
,
Li
Y
, et al
.
Outcomes after reinfusion of CD19-specific chimeric antigen receptor (CAR)-modified T cells in children and young adults with relapsed/refractory B-cell acute lymphoblastic leukemia
.
Blood
.
2021
;
138
(
suppl 1
):
474
.
79.
Li
AM
,
Hucks
GE
,
Dinofia
AM
, et al
.
Checkpoint inhibitors augment CD19-directed chimeric antigen receptor (CAR) T cell therapy in relapsed B-cell acute lymphoblastic leukemia
.
Blood
.
2018
;
132
(
suppl 1
):
556
.
80.
Barredo
JC
,
Devidas
M
,
Lauer
SJ
, et al
.
Isolated CNS relapse of acute lymphoblastic leukemia treated with intensive systemic chemotherapy and delayed CNS radiation: a pediatric oncology group study
.
J Clin Oncol
.
2006
;
24
(
19
):
3142
-
3149
.
81.
Rau
RE
,
Dai
Y
,
Devidas
M
, et al
.
Prognostic impact of minimal residual disease at the end of consolidation in NCI standard-risk B-lymphoblastic leukemia: a report from the Children's Oncology Group
.
Pediatr Blood Cancer
.
2021
;
68
(
4
):
e28929
.
82.
Meyr
F
,
Escherich
G
,
Mann
G
, et al
.
Outcomes of treatment for relapsed acute lymphoblastic leukaemia in children with Down syndrome
.
Br J Haematol
.
2013
;
162
(
1
):
98
-
106
.
83.
Ghorashian
S
,
Jacoby
E
,
De Moerloose
B
, et al
.
Tisagenlecleucel therapy for relapsed or refractory B-cell acute lymphoblastic leukaemia in infants and children younger than 3 years of age at screening: an international, multicentre, retrospective cohort study
.
Lancet Haematol
.
2022
;
9
(
10
):
e766
-
e775
.

Author notes

R.M.M. and N.N.S. contributed equally to this study.

Sign in via your Institution