Significant improvements have occurred for adolescent and young adult (AYA) B-cell acute lymphoblastic leukemia (B-ALL) patients following the widespread adoption of “pediatric-inspired” treatment regimens for AYA patients cared for in adult oncology settings. However, for AYA patients, aged 15 to 39, an outcomes gap remains in B-ALL, necessitating the incorporation of novel therapies into up-front treatment regimens. As a result, clinical trial enrollment remains the current standard of care for AYA B-ALL across disease subtypes when available and accessible. Currently, several up-front trials are looking to incorporate the use of inotuzumab, blinatumomab, and chimeric antigen receptor T-cell therapy into existing chemotherapy backbones for AYA patients, as well as tyrosine kinase inhibitors for both Philadelphia-positive (Ph+) and Ph-like B-ALL. In addition to ongoing attempts to improve up-front treatments by incorporating immunotherapy and targeted approaches, the increased use of next generation sequencing for measurable residual disease evaluation has led to superior risk-stratification and a decreased need to pursue consolidative hematopoietic stem cell transplantation during the first complete remission for many patients.

Learning Objectives

  • Evaluate pediatric and “pediatric-inspired” regimens for the treatment of AYA B-ALL

  • Compare current clinical trial approaches for AYA B-ALL subtypes in the up-front setting

  • Discuss changing standards for MRD assessment and the impact on consolidative HSCT in first CR

A 20-year-old man presents to an urgent care clinic for bruising and profound fatigue. Blood work shows anemia, thrombocytopenia, and a white blood cell count (WBC) of 32 × 109/L. Since he is over 18 years old, the patient is referred to the adult emergency room, where a repeat complete blood count shows 68% lymphoblasts on manual differential with morphology suggestive of ALL (acute lymphoblastic leukemia). The patient is admitted to the Adult Leukemia Service for a further work-up and initiation of treatment.

Up-front treatment for adolescent and young adult (AYA) patients with B-cell ALL (B-ALL) at pediatric centers consists of a 4-drug induction, augmented Berlin-Frankfurt-Münster (BFM) consolidation, interim maintenance with high-dose methotrexate, delayed intensification, a second interim maintenance with Capizzi methotrexate, and, finally, an extended maintenance therapy (Figure 1). This treatment backbone has experienced only minor changes over the last few decades, including the shortening of maintenance to 2 years for males and a decrease in frequency of steroid/vincristine pulses to every 12 weeks.1  These modifications were adapted for the Children's Oncology Group (COG) current up-front high-risk trial AALL1732 (NCT03959085). While variations on the COG backbone exist, like the Associazone Italiana Ematologia Oncologia Pediatrica-BFM (eg, AIEOP-BFM ALL 2017, NCT03643276) or St Jude Total Therapy (eg, St Jude Total XVII, NCT03117751), many of the same general treatment principles persist. Compared to adult ALL regimens, pediatric approaches are less myelosuppressive and include more asparaginase with greater total doses of steroids, vincristine, and intrathecal chemotherapy.2  Despite the ability of many pediatric hospitals to treat older adolescent B-ALL patients, even up to the age of 21 to 24 years, the majority of AYA patients over 18 (87.1%) are managed by adult oncologists, often in community settings, which has traditionally limited exposure to pediatric regimens.3 

Figure 1.

Up-front AYA B-ALL treatment schemas. Ara-C, cytarabine; CNS3, central nervous system; CPM, cyclophosphamide; cXRT, cranial radiation; DEX, dexamethasone; DNR, daunorubicin; DOX, doxorubicin; HD-MTX, high-dose methotrexate; IT-MTX, intrathecal methotrexate; IT-Arac, intrathecal cytarabine; PEG, pegaspargase; PO-MTX, oral methotrexate; Pred, prednisone; 6-MP, mercaptopurine; 6-TG, thioguanine; VCR, vincristine.

Figure 1.

Up-front AYA B-ALL treatment schemas. Ara-C, cytarabine; CNS3, central nervous system; CPM, cyclophosphamide; cXRT, cranial radiation; DEX, dexamethasone; DNR, daunorubicin; DOX, doxorubicin; HD-MTX, high-dose methotrexate; IT-MTX, intrathecal methotrexate; IT-Arac, intrathecal cytarabine; PEG, pegaspargase; PO-MTX, oral methotrexate; Pred, prednisone; 6-MP, mercaptopurine; 6-TG, thioguanine; VCR, vincristine.

Close modal

Following a large retrospective study showing a doubling of survival for AYA patients treated on US pediatric cooperative group trials compared to US adult cooperative group trials (7-year event-free survival [EFS], 63% vs 34%; P < .001),4  a prospective phase 2 trial evaluated the use of a “pediatric-inspired” regimen by adult oncology providers. Based on the pediatric AALL0232 backbone, CALGB10403 showed both an improved EFS of 59% and overall survival (OS) of 73% when compared to historical controls. The regimen also had acceptable toxicity rates in the treatment population, and delivery was feasible in the adult oncology setting.5  Superior outcomes using pediatric- inspired regimens have also been demonstrated across multiple prospective trials by other cooperative groups (Table 1),2  resulting in the pediatric-inspired approach becoming the new standard of care for AYA B-ALL patients treated in adult centers.6 

Table 1.

“Pediatric-inspired” B-ALL trials

TrialNo. of patientsAge (years), median (range)CR (%)OS (%)EFS (%)DFS (%)Duration of follow-upAlloHSCT
JALSG 202-U 139 19 (16-24) 97 74 71 4 year t(4;11) 
DFC1 01-1756 92 28 (18-50) 86 70 71 4 year t(4;11), +8, Ph+ 
DFCI 06-254 110 32 (18-50) 89 75 73 3 year t(4;11), +8, Ph+ 
UKALL 2003 229 16-24 97 76.4 72.3 5 year  
MDACC aBFM 106 22 (13-39) 93 53 60 5 year t(4;11) or MRD+ 
NOPHO ALL 2008 221 26 (18-45) 78 74 5 year D29 MRD >5% or D79 > 0.1% 
GRAALL 2005 787 36 (18-60) 92 58.5 52 5 year High risk or MRD+ 
CALGB10403 295 24 (17-39) 89 73 59 66 3 year  
ALLRE08 PETHEMA 89 20 (15-29) 95 74 62 65 5 year MRD+ 
ALL06 86 22 (15-39) 90.2 74.9 72.8 3 year t(4;11), MRD+, poor steroid response or WBC >100 
GIMEMA LAL-1308 76 23 (18-35) 92 60.3 60.4 4 year No CR at D33, pro-B ALL or WBC >100, t(4;11), MRD+ 
TrialNo. of patientsAge (years), median (range)CR (%)OS (%)EFS (%)DFS (%)Duration of follow-upAlloHSCT
JALSG 202-U 139 19 (16-24) 97 74 71 4 year t(4;11) 
DFC1 01-1756 92 28 (18-50) 86 70 71 4 year t(4;11), +8, Ph+ 
DFCI 06-254 110 32 (18-50) 89 75 73 3 year t(4;11), +8, Ph+ 
UKALL 2003 229 16-24 97 76.4 72.3 5 year  
MDACC aBFM 106 22 (13-39) 93 53 60 5 year t(4;11) or MRD+ 
NOPHO ALL 2008 221 26 (18-45) 78 74 5 year D29 MRD >5% or D79 > 0.1% 
GRAALL 2005 787 36 (18-60) 92 58.5 52 5 year High risk or MRD+ 
CALGB10403 295 24 (17-39) 89 73 59 66 3 year  
ALLRE08 PETHEMA 89 20 (15-29) 95 74 62 65 5 year MRD+ 
ALL06 86 22 (15-39) 90.2 74.9 72.8 3 year t(4;11), MRD+, poor steroid response or WBC >100 
GIMEMA LAL-1308 76 23 (18-35) 92 60.3 60.4 4 year No CR at D33, pro-B ALL or WBC >100, t(4;11), MRD+ 

alloHCT, allogeneic hematopoietic stem cell transplantation; MRD, measurable residual disease.

Adapted from Carobolante et al.2 

Alternative AYA treatment approaches do exist, including MD Anderson Cancer Center's hyperCVAD regimen (Figure 1), as well as its augmented hyperCVAD that adds blocks of pegylated asparaginase. This regimen was shown in its single-center retrospective analysis to produce similar results to the BFM-based approach in AYA patients.7  However, in other retrospective analyses it was found to be inferior compared to pediatric-inspired regimens in regard to 3-year OS (48.5% vs 72.6%; P = .4), mean OS (41.5 ± 6.4 months vs 53.9 ± 5.4 months; P = .012), mean relapse-free survival (RFS; 39.1 ± 6.8 months vs 53.9 ± 5.4 months; P = .009), and 3-year disease-free survival (DFS; 54.7% vs 76.4%; P = .44).8  An additional comparison of 2 pediatric-inspired regimens with hyperCVAD found superior complete remission (CR) rates (79.5% vs 64.2%; P = .02), lower relapse rates (44.1% vs 60.0%; P = .04), and improved 24-month OS (41.5% vs 28.1%; P = .012) with the pediatric-inspired regimens. Treatment per CALGB10403 was also the only independent prognostic factor for OS in patients older than 20 years (hazard ratio, 0.44; 95% CI, 0.20-0.97; P = .04).9 

Following admission to the Adult Leukemia Service, the patient is confirmed on multiparameter flow cytometry (MFC) to have B-ALL that is CD20+. He is initiated on treatment per CALGB10403 with fluorescence in situ hybridization, mutational profile, and next generation sequencing (NGS) testing pending.

Regardless of the up-front treatment utilized in the AYA population (Figure 1), a survival gap exists in AYA B-ALL when compared to pediatric outcomes. Following up-front therapy, children aged 1 to 14 years now achieve a 5-year OS of greater than 93%, whereas the OS for AYA patients ranges from 60% to 78% despite CR rates of 85% to 95%.10  The cause of this outcome difference is multifactorial, including nonrelapse mortality from an increased risk of treatment-related toxicities compared to pediatric patients and the prevalence of chemoresistant disease due to the particular B-ALL mutational profile seen in AYA patients.11,12  Among early efforts to improve B-ALL outcomes was the introduction of the CD20 monoclonal antibody rituximab to up-front therapy based on the GRAALL-2005/R randomized trial showing improved EFS and decreased rates of relapse in CD20+ adult B-ALL patients.13  This is not standard practice in pediatric regimens, which do not incorporate rituximab into up-front treatment due to a lack of significant difference in measurable residual disease (MRD) response rates in patients receiving rituximab and those who do not receive it.14 

Currently, given the emergence of novel agents (Figure 2) and the success of inotuzumab, blinatumomab, and CD19 chimeric antigen receptor (CAR) T cells in the relapsed/refractory (R/R) B-ALL setting,15  the preferred up-front treatment for Ph− B-ALL is clinical trial enrollment, when available and accessible, investigating the incorporation of these treatment modalities.

Figure 2.

Time line of treatment approvals in B-ALL. Brexu-cel, brexucabtagene autoleucel; Car, chimeric antigen receptor; MRD, measurable residual disease; Ph-neg, Philadelphia negative; R/R, relapsed/refractory; Tisa-cel, tisagenlecleucel.

Figure 2.

Time line of treatment approvals in B-ALL. Brexu-cel, brexucabtagene autoleucel; Car, chimeric antigen receptor; MRD, measurable residual disease; Ph-neg, Philadelphia negative; R/R, relapsed/refractory; Tisa-cel, tisagenlecleucel.

Close modal

Inotuzumab

For AYA patients treated in pediatric centers, the up-front COG trial AALL1732 randomizes Ph− B-ALL patients who are end of consolidation (EOC) MRD-negative to receive 2 cycles of the anti-CD22 antibody drug conjugate inotuzumab (NCT03959085). A comparable study at adult centers is the successor to CALGB10403, Alliance trial A041501, which is investigating the use of 2 cycles of inotuzumab postinduction for MRD eradication (NCT03150693). Patients aged 18 to 25 would be eligible for either trial depending on treatment location.

Inotuzumab was chosen for these trials based on the results of the adult phase 3 INO-VATE trial showing both an improved CR rate (73.8% vs 30.9%; P < .0001) and MRD-negative CR (78.4% vs 28.1%; P < .001) in R/R B-ALL compared to chemotherapy,16  as well as an acceptable safety profile from the R/R pediatric B-ALL trial COG AALL1621.17  While the most frequent grade 3 adverse event in prior inotuzumab trials was hepatic toxicity, including cases of veno-occlusive disease, both AALL1732 and A041501 have noted increased rates of sepsis during subsequent blocks of chemotherapy, requiring protocol modifications.18  Although not temporally associated with inotuzumab infusion, concern exists for both prolonged myelosuppression with chemotherapy following inotuzumab exposure and inotuzumab-induced hypogammaglobinemia leading to increased risk of bacterial infection.

In addition to AALL1732 and A041501, numerous ongoing trials including AYA patients are also exploring the role of inotuzumab as part of postinduction therapy and its efficacy for MRD eradication in up-front therapy (Table 2).

Table 2.

Up-front inotuzumab and blinatumomab trials

TrialPlanned patientsAgesInterventionStatusClinical trials
Inotuzumab      
 COG AALL1732 4772 1-25 2 cycles of Ino in consolidation Recruiting NCT03959085 
 ALLIANCE A041501 310 18-39 2 cycles of Ino in consolidation Suspended NCT03150693 
 ALLTogether 6430 0-45 2 cycles of Ino prior to maintenance Recruiting NCT04307576 
 GRAALL (B-2022) 600 18-65 1 cycle of Ino in delayed intensification Planned Pending9  
Blinatumomab      
 Jiangsu Institute of Hematology 35 15-59 Reduced-intensity induction + blina Recruiting NCT05557110 
 HOVON146ALL 71 18-70 Steroid + blina prephase; blina for 2 cycles in consolidation Closed to accrual NCT03541083 
 Blina-CELL (CZECRIN) 45 18-65 1 cycle of blina + chemo for induction Closed to accrual NCT04554485 
 GIMEMA (LAL2317) 149 18-65 2 cycles of blina added to chemo Closed to accrual NCT03367299 
 GRAALL (B/2014-QUEST) 95 18-59 5 cycles of blina added to consolidation/ maintenance for MRD+ Closed to accrual NCT03709719 
 GMALL (MolAct-1) 84 18+ Up to 2 cycles of blina added to chemo Completed, results pending NCT03109093 
Inotzumab + blinatumomab      
 MDACC 80 14+ Blina cycles 5-8 + Ino in cycles 6 and 8 Recruiting NCT02877303 
TrialPlanned patientsAgesInterventionStatusClinical trials
Inotuzumab      
 COG AALL1732 4772 1-25 2 cycles of Ino in consolidation Recruiting NCT03959085 
 ALLIANCE A041501 310 18-39 2 cycles of Ino in consolidation Suspended NCT03150693 
 ALLTogether 6430 0-45 2 cycles of Ino prior to maintenance Recruiting NCT04307576 
 GRAALL (B-2022) 600 18-65 1 cycle of Ino in delayed intensification Planned Pending9  
Blinatumomab      
 Jiangsu Institute of Hematology 35 15-59 Reduced-intensity induction + blina Recruiting NCT05557110 
 HOVON146ALL 71 18-70 Steroid + blina prephase; blina for 2 cycles in consolidation Closed to accrual NCT03541083 
 Blina-CELL (CZECRIN) 45 18-65 1 cycle of blina + chemo for induction Closed to accrual NCT04554485 
 GIMEMA (LAL2317) 149 18-65 2 cycles of blina added to chemo Closed to accrual NCT03367299 
 GRAALL (B/2014-QUEST) 95 18-59 5 cycles of blina added to consolidation/ maintenance for MRD+ Closed to accrual NCT03709719 
 GMALL (MolAct-1) 84 18+ Up to 2 cycles of blina added to chemo Completed, results pending NCT03109093 
Inotzumab + blinatumomab      
 MDACC 80 14+ Blina cycles 5-8 + Ino in cycles 6 and 8 Recruiting NCT02877303 

Blina, blinatumomab; Ino, inotuzumab; MRD+, MRD-positive.

Blinatumomab

In addition to inotuzumab, the CD3-CD19 bispecific T-cell engager blinatumomab is being evaluated in the up-front treatment setting based on its efficacy in R/R (TOWER19 ) and MRD-positive (BLAST20 ) adult B-ALL trials. Unlike inotuzumab, blinatumomab response and outcomes are superior with low pretreatment tumor burden.19,20  This has led to its superior efficacy as a consolidative treatment rather than salvage therapy, as illustrated by the results of 2 pediatric phase 3 trials in patients up to the age of 30 years.21,22  Based on the initial results of the randomized phase 3 ECOG-ACRIN E1910 trial (which included patients as young as 30 years), the standard of care going forward for patients achieving an MRD-negative remission following induction will likely include the incorporation of blinatumomab.

Utilizing a BFM-based regimen adapted from the E2993/ UKALLXII that incorporated an extended remission induction, rituximab for CD20+ patients, and pegaspargase for patients younger than 55 years, E1910 randomized patients achieving MRD-negativity at the end of induction (EOI) to receive 4 cycles of blinatumomab. With a median follow-up of 43 months, a significant improvement in median OS was seen in the blinatumomab arm (not reached vs 71.4 months; P = .003).23  Currently, only AYA patients with Down syndrome are included in the up-front pediatric blinatumomab trial AALL1731 (NCT03914625), but several other ongoing trials are assessing the efficacy of blinatumomab with or without the addition of inotuzumab in the up-front setting (Table 2).

CAR T-cell products

Similar to blinatumomab, the 2 US Food and Drug Administration (FDA)–approved CAR T-cell products for R/R B-ALL, tisagenlecleucel (tisa-cel) and brexucabtagene autoleucel (brexu-cel), create a cytotoxic T-cell response against CD19-expressing B-ALL cells, resulting in high rates of MRD-negative CR (Table 3).24  Tisa-cel is FDA approved for patients up to 25 years of age with refractory disease or those in a second or later relapse. It is being evaluated in the up-front setting for AYA patients younger than 25 years who are EOC-positive as part of COG AALL1721 (NCT03876769). The use of CAR T cells earlier in therapy may be more efficacious at this time point given the effects of high disease burden and greater number of prior lines of therapy on outcomes.25  Tisa-cel, which utilizes a 4-1BB costimulatory domain, also has the potential for durable remission with a single CAR T-cell infusion.26,27  Based on the ability of NGS MRD assessment and loss of B-cell aplasia to predict relapse post CAR T cell, the proposed CAR-CURE BMT-CTN trial will seek to establish if CAR therapy alone is sufficient or whether a subset of patients may benefit from post-CAR consolidative hematopoietic stem cell transplantation (HSCT) (NCT05621291). Currently, no equivalent up-front trials exist for brexu-cel, which utilizes a CD28 costimulatory domain,28  or other CAR T-cell products.

Table 3.

CAR T-cell therapies FDA approved for AYA B-ALL

TrialNo. of patientsMedian age (range), yearsCR/CRi (%)MRD-negativeCR in responders (%)Median RFS (mo)Median OS (mo)Unique toxicity
Tisa-cel        
 ELIANA26  75 11 (3-23) 81 100 EFS at 12 months = 50% 19.1 CRS (any): 77%
Gr. ≥3: 35%
ICANS (any): 40%
Grade ≥3: 13% 
Brexu-cel        
 ZUMA-3, phase 1/228  78 42.5 (18-84) 73 97 11.7 25.4 CRS (any): 93%
Gr. ≥3: 31%
ICANS (any): 78%
Gr. ≥3: 38% 
TrialNo. of patientsMedian age (range), yearsCR/CRi (%)MRD-negativeCR in responders (%)Median RFS (mo)Median OS (mo)Unique toxicity
Tisa-cel        
 ELIANA26  75 11 (3-23) 81 100 EFS at 12 months = 50% 19.1 CRS (any): 77%
Gr. ≥3: 35%
ICANS (any): 40%
Grade ≥3: 13% 
Brexu-cel        
 ZUMA-3, phase 1/228  78 42.5 (18-84) 73 97 11.7 25.4 CRS (any): 93%
Gr. ≥3: 31%
ICANS (any): 78%
Gr. ≥3: 38% 

CRi, complete remission with incomplete count recovery; CRS, cytokine release syndrome; Gr, grade, ICANS, immune effector cell-associated neurotoxicity syndrome.

Compared to pediatric B-ALL, where it accounts for 1% to 2% of cases, BCR-ABL1 rearrangement or Philadelphia-positive (Ph+) B-ALL is the most frequent genetic subcategory in adults, including an incidence in young adult patients of approximately 25% to 30%. Although previously carrying a dismal prognosis with an OS of less than 25% in adults and less than 50% in pediatric patients, the introduction of tyrosine kinase inhibitors (TKIs) has significantly altered overall outcomes.29  Ongoing trials including AYA patients seek to understand both the ideal TKI selection and overall treatment intensity, as well as the need for postremission HSCT following the incorporation of third-generation TKIs and/or blinatumomab into up-front therapy.

Despite remission rates of 95% to 100% with the integration of TKIs into pediatric-inspired or hyperCVAD regimens,29  relapse is common and is associated with kinase domain mutations that can be detected at the time of diagnosis.30  Compared to the first- and second-generation TKIs, ponatinib can overcome mutations like T315I and was shown to have a 3-year OS superior to dasatinib when combined with hyperCVAD for frontline therapy (83% vs 56%; P = .03).31  While it may become the preferred TKI in adult B-ALL, ponatinib is currently used in pediatrics only for relapsed disease or if a resistance mutation is detected. The efficacy of ponatinib in younger patients is being tested in the R/R setting as part of the phase 1/2 COG trial AALL1922, given the lack of pharmacokinetic and safety data for patients less than 18 years (NCT04501614).

Similar to Ph− B-ALL, current up-front trials are incorporating blinatumomab in combination with TKIs in hopes of improving EFS/OS and allowing chemotherapy dose reduction (Table 4).

Table 4.

Newly diagnosed Ph+ and Ph-like clinical trials

TrialPlanned patientsAgesInterventionStatusClinical trials
Ph+      
 EsPhALL2017/
 COG AALL1631 
475 1-21 Standard risk: imatinib with EsPhALL vs COG high-risk pre-B ALL backbone
High risk: alloBMT with imatinib maintenance 
Recruiting NCT03007147 
 EA9181 330 18-75 Dasatinib or ponatinib/blinatumomab vs dasatinib or ponatinib/Hyper-CVAD Recruiting NCT04530565 
 GIMEMA
 ALL2820 
236 18+ Ponatinib/blinatumomab vs imatinib/ chemotherapy Recruiting NCT04722848 
 U Chicago 25 18+ Inotuzumab + dasatinib + steroid Induction Recruiting NCT04747912 
Ph-like      
 COG AALL1131 22 1-31 Dasatanib with chemotherapy for ABL-class fusions Closed to accrual NCT02883049 
 COG AALL1521 171 1-21 Ruxolitinib with chemotherapy for CRLF2 rearrangements and other JAK pathway alterations Closed to accrual NCT02723994 
 Total Therapy XVII 790 1-18 Dasatinib: patients with Ph+ and those with ABL-class fusion
Ruxolitinib: patients with activation of JAK/STAT signaling 
Closed to accrual NCT03117751 
 UCCC 1920 15 18-39 Ruxolitinib: patients with activation of JAK/STAT signaling Recruiting NCT03571321 
TrialPlanned patientsAgesInterventionStatusClinical trials
Ph+      
 EsPhALL2017/
 COG AALL1631 
475 1-21 Standard risk: imatinib with EsPhALL vs COG high-risk pre-B ALL backbone
High risk: alloBMT with imatinib maintenance 
Recruiting NCT03007147 
 EA9181 330 18-75 Dasatinib or ponatinib/blinatumomab vs dasatinib or ponatinib/Hyper-CVAD Recruiting NCT04530565 
 GIMEMA
 ALL2820 
236 18+ Ponatinib/blinatumomab vs imatinib/ chemotherapy Recruiting NCT04722848 
 U Chicago 25 18+ Inotuzumab + dasatinib + steroid Induction Recruiting NCT04747912 
Ph-like      
 COG AALL1131 22 1-31 Dasatanib with chemotherapy for ABL-class fusions Closed to accrual NCT02883049 
 COG AALL1521 171 1-21 Ruxolitinib with chemotherapy for CRLF2 rearrangements and other JAK pathway alterations Closed to accrual NCT02723994 
 Total Therapy XVII 790 1-18 Dasatinib: patients with Ph+ and those with ABL-class fusion
Ruxolitinib: patients with activation of JAK/STAT signaling 
Closed to accrual NCT03117751 
 UCCC 1920 15 18-39 Ruxolitinib: patients with activation of JAK/STAT signaling Recruiting NCT03571321 

alloBMT, allogeneic bone marrow transplantation.

Philadelphia-like (Ph-like) B-ALL expresses a gene signature that is quite similar to Ph+ B-ALL but portends significantly worse outcomes due to its increased resistance to asparaginase and daunorubicin and poor sensitivity to glucocorticoids.32  Ph-like B-ALL is more prevalent in AYA patients (25-30%) compared to children (10-15%), with an increased prevalence in the Hispanic/Latino and Native American population.33-35  Patients with Ph-like disease have high rates of persistent MRD-positivity and OS rates of only 20% to 30%, necessitating HSCT for long-term remissions.36  Ongoing efforts to improve outcomes in AYA patients with Ph-like B-ALL have focused on targeting the JAK/STAT signaling pathway and ABL-class fusions (Table 3) and/or incorporating immunotherapy approaches.

Phase 2 studies exploring the efficacy of incorporating the JAK inhibitor ruxolitinib into induction regimens for Ph-like ALL are still ongoing (NCT03117751 and NCT03571321), including COG AALL1521 (NCT02723994) for patients with CRLF2 rearrangements or JAK pathway mutations. Although ruxolitinib in combination with a traditional chemotherapy backbone has been shown to be safe and tolerable, it is still unknown whether outcomes are better than historical controls.32  A trial adding ruxolitinib to hyperCVAD was terminated early due to low accrual and lack of efficacy (NCT02420717).

For Ph-like patients with ABL rearrangements, prior reports have suggested the benefit of BCR-ABL–specific TKIs for patients with rearrangements of PDGFRB, which are associated with a high risk of induction failure.37  COG AALL1131 (NCT02883049) was amended to test the benefit of dasatinib in patients with ABL class fusions, but results are pending.34  An interim data analysis of MDACC's phase 1/2 trial of hyper-CVAD plus dasatinib showed safety and efficacy in R/R, suggesting overall tolerability of the regimen.38 

Given the high response rates in R/R Ph-like B-ALL with inotuzumab, blinatumomab, and CAR T,39  AYA patients are also eligible for the up-front Ph− trials (Table 2).

Fluorescence in situ hybridization testing reveals Ph+ B-ALL, and the patient is found to be IKFZpos on mutational profiling. Dasatinib is added to his induction therapy as well as rituximab for CD20+ disease. His EOI MRD testing on day 29 is positive by BCR-ABL polymerase chain reaction (PCR) and NGS.

MRD monitoring and treatment modification

Regardless of B-ALL subtype or frontline regimen used, the most important prognostic factor for long-term outcomes in ALL remains the persistence of MRD following treatment.40  While acceptable MRD testing should consist of a standardized, validated assay with a sensitivity of 10−4, the optimal time point (EOI vs EOC, or prior to HSCT) and preferred method (MFC, reverse transcriptase PCR, or NGS) remains a point of debate.41 

Compared to pediatric patients, the GRAALL-2003 and 2005 trials showed a delayed rate of MRD clearance by MFC in adult B-ALL patients. While only 36% of patients achieved MRD- negative CR at EOI, 76% of patients were in MRD-negative CR by the EOC.42  Despite the potential for later MRD clearance, postinduction MRD-positivity in Ph− AYA patients treated on CALGB10403 was an independent predictor of OS, suggesting that earlier MRD clearance may be more important for durable remissions in AYA patients.5  Currently, for both pediatric and adult treatment approaches for Ph− B-ALL, end consolidation MRD- positivity necessitates treatment modification and remains an indication for HSCT in patients able to obtain a first CR.43,44  Future studies incorporating up-front cellular and immunotherapy approaches may redefine the acceptable timing of MRD clearance.

In addition to the need to determine the importance of when MRD clears, there has been a shift from MFC to utilizing the more sensitive immunoglobulin or T-cell receptor clonotype based NGS, which has a sensitivity of up to 10−6. A recent analysis of discordant MFC and NGS outcomes showed a 5-year cumulative incidence of relapse of 0% in patients MRD-negative by both NGS and MFC compared to 36% in MRD-negative by MFC only. Patients found to be NGS negative at CR had a 5-year OS of 89% compared to 63% for NGS-positive patients, and achieving NGS MRD-negativity at a later time point did not predict low relapse risk.45  For Ph+ ALL, NGS may be superior to reverse transcriptase PCR and allow identification of patients with detectable BCR-ABL1 but low risk for relapse.46  In regard to HSCT, NGS-based MRD both pre- and post-HSCT has been shown to be predictive of transplant outcomes with a high concordance rate between peripheral blood and bone marrow sampling.47 

The introduction of pediatric-inspired regimens, the use of second- and third-generation TKIs for Ph+ disease, and the advent of improved MRD monitoring for treatment response have created a shift away from HSCT in the first CR (CR1) for the many AYA B-ALL patients. This is more in line with the pediatric approach where consolidative HSCT in CR1 is no longer indicated for several previously defined high-risk subtypes—hypodiploid ALL, Ph+, IKZFplus—if patients achieve an MRD-negative CR.48 

While once considered the standard of care for Ph− AYA patients based on results from the MRC UKALL XII/ECOG E2993 trial,49  recent retrospective analysis comparing continued treatment per CALGB10403 to consolidative HSCT in CR1 showed a superior 5-year OS (66% chemo vs 47% HCT; P < .001) and DFS (58% chemo vs 44% HCT; P < .004) with chemotherapy alone.50  HSCT also had higher nonrelapse mortality (29% HCT vs 8% chemo; P < .001), and a separate analysis found HSCT to be the only factor associated with decreased OS on multivariable analysis.51 

For Ph+ B-ALL, HSCT is no longer the standard of care for pediatric patients with chemotherapy plus TKI alone and has shown to be noninferior to HSCT.52  While transplant remains a recommendation for AYA patients in CR1 with Ph+ B-ALL with a suitable donor in adult settings,53  the addition of second- or third-generation TKIs to intensive chemotherapy can produce deep molecular responses for which HSCT has not been shown to provide additional benefit.31  For those patients proceeding to HSCT, maintenance therapy with a TKI remains the standard of care, although its benefit remains unknown with EsPhALL2017/COGAALL1631 (NCT03007147) prospectively evaluating the feasibility, toxicity, and outcomes of post-HCT TKI maintenance in the pediatric and young adult population.

Currently, proceeding to HSCT for AYA patients in CR1 is predominately reserved for patients with specific high-risk mutations (ie, Ph-like and KM2TAr) or individuals with persistent MRD with induction therapy able to achieve a remission.

The patient's TKI is switched to ponatinib, and he achieves a complete molecular remission at the EOC evaluation with negative MRD by MFC and NGS. He does not proceed to HSCT in CR1, and the patient remains in an ongoing remission during maintenance therapy with serial monitoring by BCR-ABL PCR.

Significant advances in the up-front treatment of AYA B-ALL have occurred with the adoption of pediatric-inspired regimens as a standard of care. Hope remains to further close the survival gap with the incorporation of immunotherapy and targeted agents in the up-front setting. Time will tell if the current, ongoing up-front trials in combination with more sensitive MRD testing can further improve OS/EFS, avoid the need for HSCT in CR1 for the majority of AYA patients, and lead to decreased treatment-associated toxicity through further dose reductions of chemotherapy.

Despite this optimism with improved treatment approaches, AYA B-ALL patients remain affected by significant financial and treatment-related toxicities that may lead to excessive long-term mortality and the negation of improvement in 5-year OS.54-57  Outcomes also remain disproportionately worse for minority patients,11  and substantial barriers remain for AYA clinical trial enrollment.58  In addition to improved up-front treatment regimens, attention must remain on the unique needs of this patient population, which often remain underrecognized or overlooked.59 

John C. Molina: no competing financial interests to declare.

Seth Rotz: no competing financial interests to declare.

John C. Molina: The off-label use of blinatumomab, inotuzumab ozogamicin, tisagenlecleucel, ponatinib, ruxolitinib, dasatinib is discussed.

Seth Rotz: The off-label use of blinatumomab, inotuzumab ozogamicin, tisagenlecleucel, ponatinib, ruxolitinib, dasatinib is discussed.

1.
Toksvang
LN
,
Lee
SHR
,
Yang
JJ
,
Schmiegelow
K.
Maintenance therapy for acute lymphoblastic leukemia: basic science and clinical translations
.
Leukemia
.
2022
;
36
(
7
):
1749
-
1758
.
doi:10.1038/s41375-022-01591-4
.
2.
Carobolante
F
,
Chiaretti
S
,
Skert
C
,
Bassan
R.
Practical guidance for the management of acute lymphoblastic leukemia in the adolescent and young adult population
.
Ther Adv Hematol
.
2020
;
11
(
3 February
):
204062072090353
.
doi:10.1177/2040620720903531
.
3.
Muffly
L
,
Alvarez
E
,
Lichtensztajn
D
,
Abrahao
R
,
Gomez
SL
,
Keegan
T.
Patterns of care and outcomes in adolescent and young adult acute lymphoblastic leukemia: a population-based study
.
Blood Adv
.
2018
;
2
(
8
):
895
-
903
.
doi:10.1182/bloodadvances.2017014944
.
4.
Stock
W
,
La
M
,
Sanford
B
, et al.
What determines the outcomes for adolescents and young adults with acute lymphoblastic leukemia treated on cooperative group protocols? A comparison of Children's Cancer Group and Cancer and Leukemia Group B studies
.
Blood
.
2008
;
112
(
5
):
1646
-
1654
.
doi:10.1182/blood-2008-01-130237
.
5.
Stock
W
,
Luger
SM
,
Advani
AS
, et al.
A pediatric regimen for older adolescents and young adults with acute lymphoblastic leukemia: results of CALGB 10403
.
Blood
.
2019
;
133
(
14
):
1548
-
1559
.
doi:10.1182/blood-2018-10-881961
.
6.
Siegel
SE
,
Stock
W
,
Johnson
RH
, et al.
Pediatric-inspired treatment regimens for adolescents and young adults with Philadelphia chromosome-negative acute lymphoblastic leukemia: a review
.
JAMA Oncol
.
2018
;
4
(
5
):
725
-
734
.
doi:10.1001/jamaoncol.2017.5305
.
7.
Rytting
ME
,
Jabbour
EJ
,
Jorgensen
JL
, et al.
Final results of a single institution experience with a pediatric-based regimen, the augmented Berlin-Frankfurt-Munster, in adolescents and young adults with acute lymphoblastic leukemia, and comparison to the hyper-CVAD regimen
.
Am J Hematol
.
2016
;
91
(
8
):
819
-
823
.
doi:10.1002/ajh.24419
.
8.
Siegel
SE
,
Coccia
PF
,
Barr
R
,
Hayes-Lattin
B
,
Bleyer
A.
RE: a reappraisal of sex-specific cancer survival trends among adolescents and young adults in the United States
.
JNCI: J Natl Cancer Inst
.
2019
;
111
(
6
):
633
-
634
.
doi:10.1093/jnci/djz019
.
9.
Almanza-Huante
E
,
Espinosa-Bautista
K
,
Rangel-Patiño
J
,
Demichelis- Gómez
R.
Comparison of two pediatric-inspired regimens to Hyper-CVAD in Hispanic adolescents and young adults with acute lymphoblastic leukemia
.
Clin Lymphoma Myeloma Leuk
.
2021
;
21
(
1
):
55
-
62.e2
.
doi:10.1016/j.clml.2020.07.017
.
10.
Sasaki
K
,
Jabbour
E
,
Short
NJ
, et al.
Acute lymphoblastic leukemia: a population-based study of outcome in the United States based on the surveillance, epidemiology, and end results (SEER) database, 1980-2017
.
Am J Hematol
.
2021
;
96
(
6
):
650
-
658
.
doi:10.1002/ajh.26156
.
11.
Gupta
S
,
Dai
Y
,
Chen
Z
, et al.
Racial and ethnic disparities in childhood and young adult acute lymphocytic leukaemia: secondary analyses of eight Children's Oncology Group cohort trials
.
Lancet Haematol
.
2023
;
10
(
2
):
e129
-
e141
.
doi:10.1016/S2352-3026(22)00371-4
.
12.
Boissel
N.
New developments in ALL in AYA
.
Hematology Am Soc Hematol Educ Program
.
2022
;
2022
(
1
):
190
-
196
.
doi:10.1182/hematology.2022000336
.
13.
Maury
S
,
Chevret
S
,
Thomas
X
, et al.
Rituximab in B-lineage adult acute lymphoblastic leukemia
.
N Engl J Med
.
2016
;
375
(
11
):
1044
-
1053
.
doi:10.1056/NEJMoa1605085
.
14.
Hoshitsuki
K
,
Zhou
Y
,
Miller
AM
, et al.
Rituximab administration in pediatric patients with newly diagnosed acute lymphoblastic leukemia
.
Leukemia
.
2023
;
37
(
9
):
1782
-
1791
.
doi:10.1038/s41375-023-01992-z
.
15.
Aldoss
I
,
Shah
BD
,
Park
JH
, et al.
Sequencing antigen-targeting antibodies and cellular therapies in adults with relapsed/refractory B-cell acute lymphoblastic leukemia
.
Am J Hematol
.
2023
;
98
(
4
):
666
-
680
.
doi:10.1002/ajh.26853
.
16.
Kantarjian
HM
,
DeAngelo
DJ
,
Stelljes
M
, et al.
Inotuzumab ozogamicin versus standard of care in relapsed or refractory acute lymphoblastic leukemia: final report and long-term survival follow-up from the randomized, phase 3 INO-VATE study
.
Cancer
.
2019
;
125
(
14
):
2474
-
2487
.
doi:10.1002/cncr.32116
.
17.
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
.
doi:10.1200/JCO.21.01693
.
18.
McNeer
JL
,
Rheingold
SR
,
Devidas
M
, et al.
A phase 3 trial of inotuzumab ozogamicin for high-risk B-ALL: second safety phase results from Children's Oncology Group AALL1732
.
J Clin Oncol
.
2023
;
41
(
suppl 16
):
10016
.
doi:10.1200/JCO.2023.41.16_suppl.10016
.
19.
Kantarjian
H
,
Stein
A
,
Gokbuget
N
, et al.
Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia
.
N Engl J Med
.
2017
;
376
(
9
):
836
-
847
.
doi:10.1056/NEJMoa1609783
.
20.
Gokbuget
N
,
Dombret
H
,
Bonifacio
M
, et al.
Blinatumomab for minimal residual disease in adults with B-cell precursor acute lymphoblastic leukemia
.
Blood
.
2018
;
131
(
14
):
1522
-
1531
.
doi:10.1182/blood-2017-08-798322
.
21.
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
.
doi:10.1001/jama.2021.0669
.
22.
Locatelli
F
,
Zugmaier
G
,
Rizzari
C
, et al.
Effect of blinatumomab vs chemotherapy on event-free survival among children with high-risk first-relapse B-cell acute lymphoblastic leukemia: a randomized clinical trial
.
JAMA
.
2021
;
325
(
9
):
843
-
854
.
doi:10.1001/jama.2021.0987
.
23.
Litzow
MR
,
Sun
Z
,
Paietta
E
, et al.
Consolidation therapy with blinatumomab improves overall survival in newly diagnosed adult patients with B-lineage acute lymphoblastic leukemia in measurable residual disease negative remission: results from the ECOG-ACRIN E1910 randomized phase III National Cooperative Clinical Trials Network Trial
.
Blood
.
2022
;
140
(
suppl 2
):
LBA-1
-
LBA-1
.
doi:10.1182/blood-2022-171751
.
24.
June
CH
,
Sadelain
M.
Chimeric antigen receptor therapy
.
N Engl J Med
.
2018
;
379
(
1
):
64
-
73
.
doi:10.1056/NEJMra1706169
.
25.
Myers
RM
,
Shah
NN
,
Pulsipher
MA.
How I use risk factors for success or failure of CD19 CAR T cells to guide management of children and AYA with B-cell ALL
.
Blood
.
2023
;
141
(
11
):
1251
-
1264
.
doi:10.1182/blood.2022016937
.
26.
Qayed
M
,
Bleakley
M
,
Shah
NN.
Role of chimeric antigen receptor T-cell therapy: bridge to transplantation or stand-alone therapy in pediatric acute lymphoblastic leukemia
.
Curr Opin Hematol
.
2021
;
28
(
6
):
373
-
379
.
doi:10.1097/moh.0000000000000685
.
27.
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
.
doi:10.13039/100008272
.
28.
Shah
BD
,
Ghobadi
A
,
Oluwole
OO
, et al.
Two-year follow-up of KTE-X19 in patients with relapsed or refractory adult B-cell acute lymphoblastic leukemia in ZUMA-3 and its contextualization with SCHOLAR-3, an external historical control study
.
J Hematol Oncol
.
2022
;
15
(
1
):
170
.
doi:10.1186/s13045-022-01379-0
.
29.
Wieduwilt
MJ.
Ph+ ALL in 2022: is there an optimal approach?
Hematology
.
2022
;
2022
(
1
):
206
-
212
.
doi:10.1182/hematology.2022000338
.
30.
Pfeifer
H
,
Wassmann
B
,
Pavlova
A
, et al.
Kinase domain mutations of BCR-ABL frequently precede imatinib-based therapy and give rise to relapse in patients with de novo Philadelphia-positive acute lymphoblastic leukemia (Ph+ ALL)
.
Blood
.
2007
;
110
(
2
):
727
-
734
.
doi:10.1182/blood-2006-11-052373
.
31.
Jabbour
E
,
Haddad
FG
,
Short
NJ
,
Kantarjian
H.
Treatment of adults with Philadelphia chromosome-positive acute lymphoblastic leukemia—from intensive chemotherapy combinations to chemotherapy-free regimens: a review
.
JAMA Oncol
.
2022
;
8
(
9
):
1340
-
1348
.
doi:10.1001/jamaoncol.2022.2398
.
32.
Alghandour
R
,
Sakr
DH
,
Shaaban
Y.
Philadelphia-like acute lymphoblastic leukemia: the journey from molecular background to the role of bone marrow transplant—review article
.
Ann Hematol
.
2023
;
102
(
6
):
1287
-
1300
.
doi:10.1007/s00277-023-05241-2
.
33.
Roberts
KG
,
Reshmi
SC
,
Harvey
RC
, et al.
Genomic and outcome analyses of Ph-like ALL in NCI standard-risk patients: a report from the Children's Oncology Group
.
Blood
.
2018
;
132
(
8
):
815
-
824
.
doi:10.1182/blood-2018-04-841676
.
34.
Tran
TH
,
Hunger
SP.
The genomic landscape of pediatric acute lymphoblastic leukemia and precision medicine opportunities
.
Semin Cancer Biol
.
2022
;
84
(
28
):
144
-
152
.
doi:10.1016/j.semcancer.2020.10.013
.
35.
Aldoss
I
,
Yang
D
,
Tomasian
V
, et al.
Outcomes of allogeneic hematopoietic cell transplantation in adults with fusions associated with Ph-like ALL
.
Blood Adv
.
2022
;
6
(
17
):
4936
-
4948
.
doi:10.1182/bloodadvances.2022007597
.
36.
Pui
CH
,
Roberts
KG
,
Yang
JJ
,
Mullighan
CG.
Philadelphia chromosome-like acute lymphoblastic leukemia
.
Clin Lymphoma Myeloma Leuk
.
2017
;
17
(
8
):
464
-
470
.
doi:10.1016/j.clml.2017.03.299
.
37.
Schwab
C
,
Ryan
SL
,
Chilton
L
, et al.
EBF1-PDGFRB fusion in pediatric B-cell precursor acute lymphoblastic leukemia (BCP-ALL): genetic profile and clinical implications
.
Blood
.
2016
;
127
(
18
):
2214
-
2208
.
doi:10.1182/blood-2015-09-670166
.
38.
Tasian
SK
,
Assad
A
,
Hunter
DS
,
Du
Y
,
Loh
ML.
A phase 2 study of ruxolitinib with chemotherapy in children with Philadelphia chromosome-like acute lymphoblastic leukemia (INCB18424-269/AALL1521): dose-finding results from the part 1 safety phase
.
Blood
.
2018
;
132
(
suppl 1
):
555
.
doi:10.1182/blood-2018-99-110221
.
39.
Aldoss
I
,
Afkhami
M
,
Yang
D
, et al.
High response rates and transition to transplant after novel targeted and cellular therapies in adults with relapsed/refractory acute lymphoblastic leukemia with Philadelphia-like fusions
.
Am J Hematol
.
2023
;
98
(
6
):
848
-
856
.
doi:10.1002/ajh.26908
.
40.
Berry
DA
,
Zhou
S
,
Higley
H
, et al.
Association of minimal residual disease with clinical outcome in pediatric and adult acute lymphoblastic leukemia: a meta-analysis
.
JAMA Oncol
.
2017
;
3
(
7
):
e170580
.
doi:10.1001/jamaoncol.2017.0580
.
41.
Della Starza
I
,
De Novi
LA
,
Elia
L
, et al.
Optimizing molecular minimal residual disease analysis in adult acute lymphoblastic leukemia
.
Cancers (Basel)
.
2023
;
15
(
2
):
374
.
doi:10.3390/cancers15020374
.
42.
Dhédin
N
,
Huynh
A
,
Maury
S
, et al.
Role of allogeneic stem cell transplantation in adult patients with Ph-negative acute lymphoblastic leukemia
.
Blood
.
2015
;
125
(
16
):
2486
-
2496
.
doi:10.1182/blood-2014-09-599894
.
43.
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
.
doi:10.1002/pbc.28929
.
44.
Short
NJ
,
Jabbour
E
,
Albitar
M
, et al.
Recommendations for the assessment and management of measurable residual disease in adults with acute lymphoblastic leukemia: a consensus of North American experts
.
Am J Hematol
.
2019
;
94
(
2
):
257
-
265
.
doi:10.1002/ajh.25338
.
45.
Short
NJ
,
Kantarjian
H
,
Ravandi
F
, et al.
High-sensitivity next-generation sequencing MRD assessment in ALL identifies patients at very low risk of relapse
.
Blood Adv
.
2022
;
6
(
13
):
4006
-
4014
.
doi:10.1182/bloodadvances.2022007378
.
46.
Short
NJ
,
Jabbour
E
,
Macaron
W
, et al.
Ultrasensitive NGS MRD assessment in Ph+ ALL: prognostic impact and correlation with RT-PCR for BCR::ABL1
.
Am J Hematol
.
2023
;
98
(
8
):
1196
-
1203
.
doi:10.1002/ajh.26949
.
47.
Liang
EC
,
Dekker
SE
,
Sabile
JMG
, et al.
Next-generation sequencing-based MRD in adults with ALL undergoing hematopoietic cell transplantation
.
Blood Adv
.
2023
;
7
(
14
):
3395
-
3402
.
doi:10.1182/bloodadvances.2023009856
.
48.
Truong
TH
,
Jinca
C
,
Mann
G
, et al.
Allogeneic hematopoietic stem cell transplantation for children with acute lymphoblastic leukemia: shifting indications in the era of immunotherapy
.
Front Pediatr
.
2021
;
9
(
December
):
782785
.
doi:10.3389/fped.2021.782785
.
49.
Goldstone
AH
,
Richards
SM
,
Lazarus
HM
, et al.
In adults with standard-risk acute lymphoblastic leukemia, the greatest benefit is achieved from a matched sibling allogeneic transplantation in first CR, and an autologous transplantation is less effective than conventional consolidation/ maintenance chemotherapy in all patients: final results of the International ALL Trial (MRC UKALL XII/ECOG E2993)
.
Blood
.
2008
;
111
(
4
):
1827
-
1833
.
doi:10.1182/blood-2007-10-116582
.
50.
Wieduwilt
MJ
,
Stock
W
,
Advani
A
, et al.
Superior survival with pediatric-style chemotherapy compared to myeloablative allogeneic hematopoietic cell transplantation in older adolescents and young adults with Ph-negative acute lymphoblastic leukemia in first complete remission: analysis from CALGB 10403 and the CIBMTR
.
Leukemia
.
2021
;
35
(
7
):
2076
-
2085
.
doi:10.1038/s41375-021-01213-5
.
51.
Seftel
MD
,
Neuberg
D
,
Zhang
MJ
, et al.
Pediatric-inspired therapy compared to allografting for Philadelphia chromosome-negative adult ALL in first complete remission
.
Am J Hematol
.
2016
;
91
(
3
):
322
-
329
.
doi:10.1002/ajh.24285
.
52.
Schultz
KR
,
Carroll
A
,
Heerema
NA
, et al.
Long-term follow-up of imatinib in pediatric Philadelphia chromosome-positive acute lymphoblastic leukemia: Children's Oncology Group study AALL0031
.
Leukemia
.
2014
;
28
(
7
):
1467
-
1471
.
doi:10.1038/leu.2014.30
.
53.
Brown
PA
,
Shah
B
,
Advani
A
, et al.
Acute lymphoblastic leukemia, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology
.
J Natl Compr Canc Netw
.
2021
;
19
(
9
):
1079
-
1109
.
doi:10.6004/jnccn.2021.0042
.
54.
Parsons
SK
,
Keegan
THM
,
Kirchhoff
AC
,
Parsons
HM
,
Yabroff
KR
,
Davies
SJ.
Cost of cancer in adolescents and young adults in the United States: results of the 2021 report by Deloitte Access Economics, commissioned by Teen Cancer America
.
J Clin Oncol
.
2023
;
41
(
17
):
3260
-
3268
.
doi:10.1200/JCO.22.01985
.
55.
Muffly
L
,
Maguire
FB
,
Li
Q
,
Kennedy
V
,
Keegan
TH.
Late effects in survivors of adolescent and young adult acute lymphoblastic leukemia
.
JNCI Cancer Spectr
.
2020
;
4
(
4
):
pkaa025
.
doi:10.1093/jncics/pkaa025
.
56.
Williams
AM
,
Liu
Q
,
Bhakta
N
, et al.
Rethinking success in pediatric oncology: beyond 5-year survival
.
J Clin Oncol
.
2021
;
39
(
20
):
2227
-
2231
.
doi:10.1200/JCO.20.03681
.
57.
Berkman
AM
,
Andersen
CR
,
Cuglievan
B
, et al.
Long-term outcomes among adolescent and young adult survivors of acute leukemia: a surveillance, epidemiology, and end results analysis
.
Cancer Epidemiol Biomarkers Prev
.
2022
;
31
(
6
):
1176
-
1184
.
doi:10.1158/1055-9965.EPI-21-1388
.
58.
Mittal
N
,
Saha
A
,
Avutu
V
,
Monga
V
,
Freyer
DR
,
Roth
M.
Shared barriers and facilitators to enrollment of adolescents and young adults on cancer clinical trials
.
Sci Rep
.
2022
;
12
(
1
):
3875
.
doi:10.1038/s41598-022-07703-5
.
59.
Jin
Z
,
Griffith
MA
,
Rosenthal
AC.
Identifying and meeting the needs of adolescents and young adults with cancer
.
Curr Oncol Rep
.
2021
;
23
(
2
):
17
.
doi:10.1007/s11912-020-01011-9
.