Pediatric Lymphomas and Leukemias: Treatment Approaches
Overview of Pediatric Acute Lymphoblastic Leukemia (ALL)
Pediatric ALL should be treated with risk-stratified multiagent chemotherapy at specialized cancer centers, consisting of induction, consolidation, and maintenance phases over 2-3 years, with treatment intensity determined by minimal residual disease (MRD) status and molecular/cytogenetic features. 1, 2
Current Survival Outcomes
- Pediatric ALL has achieved remarkable cure rates of approximately 89-90%, representing one of the most successful treatment paradigms in pediatric oncology 1, 3, 4
- This dramatic improvement from ~10% survival in the 1960s stems from risk-adapted multiagent chemotherapy, effective CNS prophylaxis, molecular understanding of disease biology, and MRD monitoring 3, 4
- Infants younger than 1 year remain an exception with 6-year overall survival of only 58.2%, showing no improvement over 30 years 1
- Adolescent and young adult (AYA) patients have lower survival rates (61% 5-year OS) compared to younger children, though outcomes improve when treated with pediatric-inspired regimens 1, 5
Treatment Structure and Components
Induction Therapy (Weeks 1-4)
Begin with multiagent induction chemotherapy including vincristine, corticosteroids (dexamethasone or prednisone), and asparaginase, with or without anthracycline depending on risk stratification. 1, 2, 4
- Three-drug induction (dexamethasone, asparaginase, vincristine) achieves 70-90% complete remission rates for standard-risk B-ALL 1
- Four-drug induction adding anthracycline is reserved for higher-risk patients 1
- Asparaginase is critical as it is relatively non-myelosuppressive and improves long-term survival to 30-50% when added to two-drug regimens 1, 6
Risk Stratification Based on MRD
MRD assessment after induction is the single most important prognostic factor and determines subsequent treatment intensity. 1, 2, 3
- MRD-negative patients (<0.01% blasts) proceed to standard consolidation and maintenance therapy 1, 2
- MRD-positive patients (≥0.01% blasts) require intensified consolidation with consideration of blinatumomab or tisagenlecleucel 1, 2
- Persistently positive MRD after consolidation warrants immunotherapy (blinatumomab or tisagenlecleucel) before proceeding to hematopoietic stem cell transplantation (HSCT) 1, 2
Consolidation Therapy (Months 2-6)
Consolidation includes high-dose methotrexate (for high-risk and T-ALL), cyclophosphamide, cytarabine, and repeated doses of asparaginase. 1, 4
- High-risk patients receive intermediate-dose methotrexate (1 g/m² over 24 hours) with leucovorin rescue 1
- T-ALL patients benefit from higher cyclophosphamide doses and triple intrathecal therapy 1
- Delayed intensification (re-induction) with dexamethasone, vincristine, and asparaginase improves outcomes, particularly in non-intensive protocols 1
Maintenance Therapy (Up to 2-3 Years Total)
Maintenance consists of daily oral mercaptopurine and weekly methotrexate, with periodic vincristine and corticosteroid pulses, continuing for 2-3 years from diagnosis. 1, 4, 7
- Dexamethasone during maintenance improves systemic and CNS leukemia control 1
- Pharmacogenomic testing for TPMT and NUDT15 variants guides mercaptopurine dosing to prevent toxicity 1
CNS-Directed Therapy
All patients require CNS prophylaxis with intrathecal chemotherapy (methotrexate, cytarabine, and corticosteroid) starting at diagnosis, without routine cranial irradiation. 1, 2
- Triple intrathecal therapy is mandatory for T-ALL and patients with CNS involvement at diagnosis 1
- Prophylactic cranial irradiation is not recommended due to serious long-term complications and lack of survival benefit with effective systemic and intrathecal therapy 1
- Intensified intrathecal chemotherapy prevents both CNS relapse and late bone marrow relapse 2
Special Populations and Molecular Subtypes
Philadelphia Chromosome-Positive (Ph+) B-ALL
Combine intensive chemotherapy with tyrosine kinase inhibitors (imatinib 340 mg/m²/day or dasatinib) to achieve 3-year event-free survival of 80-86%. 1, 2
- TKI therapy should begin during induction and continue throughout all treatment phases 1, 2
- HSCT in first complete remission is no longer routinely required with modern TKI-based regimens 1
BCR::ABL1-Like B-ALL
Identify kinase-activating alterations (ABL1, ABL2, PDGFRB, JAK2, CRLF2 fusions) through molecular testing and consider targeted therapy with appropriate TKIs or JAK inhibitors. 1
- This subtype comprises 15-20% of pediatric B-ALL and requires intensified therapy 1
- FISH probes for ABL1, ABL2, PDGFRB, and JAK2 rearrangements guide targeted therapy selection 1
Infant ALL (Age <12 Months)
Treat with Interfant-based chemotherapy regimens incorporating elements of both ALL and AML protocols, with intensified cytarabine and anthracycline therapy. 1, 2
- KMT2A (MLL) rearrangements occur in 70-80% of infant ALL and confer poor prognosis 1
- These patients require more intensive therapy but avoid prolonged maintenance due to different disease biology 1, 2
T-Cell ALL/Lymphoblastic Lymphoma
Use intensified chemotherapy backbones with higher cyclophosphamide doses, extended asparaginase, and consideration of bortezomib addition. 1
- Modern T-ALL protocols achieve outcomes nearly equivalent to B-ALL (5-year OS ~85%) 1
- Early T-cell precursor (ETP) ALL requires identification by flow cytometry (lacks CD5, CD8, CD1a; expresses myeloid markers) but does not necessarily require treatment modification 1
- Bortezomib added to BFM backbone chemotherapy improves event-free survival and overall survival specifically in T-lymphoblastic lymphoma 1
Pediatric Aggressive Mature B-Cell Lymphomas
Burkitt Lymphoma and Diffuse Large B-Cell Lymphoma
Treat sporadic Burkitt lymphoma and DLBCL with short-duration, high-intensity multiagent chemotherapy regimens (typically 3-6 months) rather than prolonged ALL-type therapy. 1
- These are highly aggressive but curable malignancies requiring treatment at centers with specific expertise 1
- Regimens include high-dose methotrexate, cyclophosphamide, cytarabine, and rituximab for CD20-positive disease 1
- Treatment duration is much shorter (3-6 months) compared to ALL (2-3 years) 1
Relapsed/Refractory Disease Management
Risk Stratification at Relapse
Classify relapse timing as very early (<18 months from diagnosis), early (18-36 months), or late (>36 months or >6 months off therapy), with site of relapse (bone marrow vs. extramedullary) determining treatment intensity. 1
- Very early bone marrow relapse has the worst prognosis and requires HSCT in second complete remission 1
- All T-ALL relapses are considered high-risk regardless of timing or site 1
- Isolated extramedullary relapse (CNS or testicular) still requires systemic chemotherapy to prevent subsequent bone marrow relapse 2
Immunotherapy Options
For relapsed/refractory B-ALL, use blinatumomab (bispecific T-cell engager) or tisagenlecleucel (CAR T-cell therapy) to achieve MRD-negative remission before proceeding to HSCT or as definitive therapy. 1, 2
- Blinatumomab achieves 88% complete MRD response in patients with MRD ≥10⁻³ 8
- Tisagenlecleucel produces long-term remissions without subsequent HSCT in some patients, with 3-year relapse-free survival of 52% and only 22% proceeding to HSCT 1, 2
- Inotuzumab ozogamicin (anti-CD22 antibody-drug conjugate) is an alternative for CD22-positive disease 1, 2
Reinduction Chemotherapy
Use bortezomib-based reinduction regimens for first relapse, achieving 68% second complete remission rates for B-ALL and 63-72% for early relapses. 1
- Standard reinduction includes vincristine, anthracycline, corticosteroid, and asparaginase with added bortezomib 1
- For T-ALL relapse, consider myeloid-type consolidation (ADE/MAE regimens) for patients with MRD ≥5×10⁻⁴ after induction 1
Resource-Stratified Approaches
Basic Resources
In resource-limited settings, use two-drug induction (vincristine and corticosteroid) achieving 50-70% remission with 20% cure rate, adding asparaginase if affordable to improve survival to 30-50%. 1
- Two-drug induction is less toxic, requires minimal supportive care, and reduces treatment abandonment rates 1
- Twinning programs with institutions in developed countries facilitate training and difficult case management 1
Limited Resources
Implement three-drug induction (dexamethasone, asparaginase, vincristine) with risk-adapted intensification, achieving up to 60% cure rates. 1
- Add daunorubicin and extended asparaginase for poor early responders based on day 8 peripheral blood or day 15 bone marrow evaluation 1
- Use intermediate-dose methotrexate (1 g/m² over 24 hours) for high-risk cases rather than high-dose protocols 1
Enhanced/Maximum Resources
Adopt comprehensive risk-directed protocols from major cooperative groups (COG, BFM, DFCI) featuring early intrathecal therapy, high-dose methotrexate consolidation, delayed intensification, and continuation therapy. 1
Critical Pitfalls to Avoid
- Never delay treatment at non-specialized centers: The complexity of ALL therapy with required supportive care (tumor lysis prophylaxis, infection management, transfusion support) necessitates treatment at specialized cancer centers 1
- Never proceed to HSCT with detectable MRD when avoidable: Additional immunotherapy or chemotherapy to achieve MRD negativity before HSCT significantly improves post-transplant outcomes 1, 2
- Never omit CNS prophylaxis: Intrathecal chemotherapy must begin at diagnosis to prevent CNS relapse, which has poor salvage rates 1, 2
- Never use chronologic age alone for treatment decisions: AYA patients (ages 15-39) benefit from pediatric-inspired regimens rather than adult protocols, yet only 21-31% treated at adult centers receive appropriate therapy 5
- Never ignore pharmacogenomic testing: TPMT and NUDT15 variants require mercaptopurine dose adjustments to prevent life-threatening myelosuppression 1