Abstract
Early successes achieved with the CD19-targeted chimeric antigen receptor (CAR) T cell product tisagenlecleucel for the treatment of paediatric B cell acute lymphoblastic leukaemia (B-ALL) led to a historic first FDA approval of a gene therapy. Widespread CAR T cell commercialization followed, along with expansion to other indications and earlier disease settings owing to clear survival benefits. However, commercial development of additional cell therapies for paediatric malignancies has stagnated, despite several products being brought to market as treatments for various haematological malignancies in adults. In contrast to the consistent efficacy achieved across B cell malignancies, CAR T cell approaches have yet to demonstrate durable activity in patients with acute myeloid leukaemia (AML), T cell acute lymphoblastic leukaemia, solid tumours and/or central nervous system cancers, with both biological factors and broader issues of development and access constraining the field. Herein, we showcase the foundational leaps achieved through the initial trials and commercialization of CAR T cell products and contextualize how these early experiences have moulded the field. We review currently approved and investigational CAR T cell therapies for paediatric and young-adult patients, including key considerations regarding safety, access and future directions. We also discuss additional immunotherapy options that guide clinical decision-making regarding optimal utilization of CAR T cells. Although clearly tolerable and efficacious, the CD19-targeted CAR T cell strategy requires ongoing refinement, and research efforts are now geared towards fully exploiting CAR T cells and other immunotherapies to improve survival with broadened access across disease states.
Key points
-
Three chimeric antigen receptor (CAR) T cell products are approved for the treatment of B cell acute lymphoblastic leukaemia (B-ALL), although only tisagenlecleucel is licensed for use in paediatric patients, and for limited indications, with a resultant reliance on this single agent for the survival gains conferred by CAR T cell therapy in paediatric B-ALL.
-
Despite high response rates in children and young adults receiving tisagenlecleucel for B-ALL, relapse rates of ~50% present an urgent need to develop and further study optimized CAR T cell constructs and approaches.
-
Risk stratification based on pre-infusion clinical characteristics can identify subsets of patients who are at high risk of CAR T cell resistance and toxicities, and thus guide optimal bridging and consolidative strategies.
-
Current CAR T cell therapy achieves excellent health-related quality of life, with low rates of treatment-related morbidity and mortality, with a tolerability profile permissive to further development of CAR T cell therapies for non-B-cell malignancies and even non-malignant diseases.
-
CAR T cells provide unique advantages over other immunotherapies, yet the optimal sequencing of these treatments, including the role of haematopoietic stem cell transplantation, remains an active area of research.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$189.00 per year
only $15.75 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to the full article PDF.
USD 39.95
Prices may be subject to local taxes which are calculated during checkout
Similar content being viewed by others
References
Maude, S. L. et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N. Engl. J. Med. 378, 439–448 (2018).
Laetsch, T. W. et al. Three-year update of tisagenlecleucel in pediatric and young adult patients with relapsed/refractory acute lymphoblastic leukemia in the ELIANA trial. J. Clin. Oncol. 41, 1664–1669 (2023).
Laetsch, T. W. et al. Patient-reported quality of life after tisagenlecleucel infusion in children and young adults with relapsed or refractory B-cell acute lymphoblastic leukaemia: a global, single-arm, phase 2 trial. Lancet Oncol. 20, 1710–1718 (2019).
Zhai, Y. et al. Comparison of blinatumomab and CAR T-cell therapy in relapsed/refractory acute lymphoblastic leukemia: a systematic review and meta-analysis. Expert Rev. Hematol. 17, 67–76 (2024).
Sun, W. et al. Outcome of children with multiply relapsed B-cell acute lymphoblastic leukemia: a therapeutic advances in childhood leukemia & lymphoma study. Leukemia 32, 2316–2325 (2018).
Chessells, J. M. et al. Long-term follow-up of relapsed childhood acute lymphoblastic leukaemia. Br. J. Haematol. 123, 396–405 (2003).
Hunger, S. P. & Raetz, E. A. How I treat relapsed acute lymphoblastic leukemia in the pediatric population. Blood 136, 1803–1812 (2020).
Ko, R. H. et al. Outcome of patients treated for relapsed or refractory acute lymphoblastic leukemia: a Therapeutic Advances in Childhood Leukemia Consortium study. J. Clin. Oncol. 28, 648–654 (2010).
Kuhlen, M. et al. Outcome of relapse after allogeneic HSCT in children with ALL enrolled in the ALL-SCT 2003/2007 trial. Br. J. Haematol. 180, 82–89 (2018).
Lund, T. C. et al. Outcomes after second hematopoietic cell transplantation in children and young adults with relapsed acute leukemia. Biol. Blood Marrow Transpl. 25, 301–306 (2019).
Reismuller, B. et al. Outcome of children and adolescents with a second or third relapse of acute lymphoblastic leukemia (ALL): a population-based analysis of the Austrian ALL-BFM (Berlin-Frankfurt-Munster) study group. J. Pediatr. Hematol. Oncol. 35, e200–e204 (2013).
Schrappe, M. et al. Outcomes after induction failure in childhood acute lymphoblastic leukemia. N. Engl. J. Med. 366, 1371–1381 (2012).
Yaniv, I. et al. Second hematopoietic stem cell transplantation for post-transplantation relapsed acute leukemia in children: a retrospective EBMT-PDWP study. Biol. Blood Marrow Transpl. 24, 1629–1642 (2018).
Eckert, C. et al. Risk factors and outcomes in children with high-risk B-cell precursor and T-cell relapsed acute lymphoblastic leukaemia: combined analysis of ALLR3 and ALL-REZ BFM 2002 clinical trials. Eur. J. Cancer 151, 175–189 (2021).
Gardner, R. A. et al. Intent-to-treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults. Blood 129, 3322–3331 (2017).
Lee, D. W. 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 385, 517–528 (2015).
O’Leary, M. C. et al. FDA approval summary: tisagenlecleucel for treatment of patients with relapsed or refractory B-cell precursor acute lymphoblastic leukemia. Clin. Cancer Res. 25, 1142–1146 (2019).
Pasquini, M. C. et al. Real-world evidence of tisagenlecleucel for pediatric acute lymphoblastic leukemia and non-Hodgkin lymphoma. Blood Adv. 4, 5414–5424 (2020).
Schultz, L. M. 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. 40, 945–955 (2022).
Rives, S. et al. Tisagenlecleucel in pediatric and young adult patients with relapsed/refractory b-cell acute lymphoblastic leukemia (B-ALL): final analyses from the ELIANA study [abstract S112]. HemaSphere 6, 13–14 (2022).
John, S. et al. Real-world data for tisagenlecleucel in patients with R/R B-ALL: subgroup analyses from the CIBMTR registry. Blood Adv. 9, 5249–5262 (2025).
Bouchkouj, N. et al. FDA approval summary: brexucabtagene autoleucel for treatment of adults with relapsed or refractory B-cell precursor acute lymphoblastic leukemia. Oncologist 27, 892–899 (2022).
Shah, B. D. et al. KTE-X19 for relapsed or refractory adult B-cell acute lymphoblastic leukaemia: phase 2 results of the single-arm, open-label, multicentre ZUMA-3 study. Lancet 398, 491–502 (2021).
US Food and Drug Administration. FDA approves obecabtagene autoleucel for adults with relapsed or refractory B-cell precursor acute lymphoblastic leukemia. fda.gov https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-obecabtagene-autoleucel-adults-relapsed-or-refractory-b-cell-precursor-acute (2024).
Roddie, C. et al. Obecabtagene autoleucel in adults with B-cell acute lymphoblastic leukemia. N. Engl. J. Med. 391, 2219–2230 (2024).
Frey, N. V. Approval of brexucabtagene autoleucel for adults with relapsed and refractory acute lymphocytic leukemia. Blood 140, 11–15 (2022).
Shah, B. D. et al. KTE-X19 anti-CD19 CAR T-cell therapy in adult relapsed/refractory acute lymphoblastic leukemia: ZUMA-3 phase 1 results. Blood 138, 11–22 (2021).
Roloff, G. W. et al. Outcomes after brexucabtagene autoleucel administered as a standard therapy for adults with relapsed/refractory B-cell ALL. J. Clin. Oncol. 43, 558–566 (2025).
Long, A. H. et al. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat. Med. 21, 581–590 (2015).
Shah, N. N. et al. Long-term follow-up of CD19-CAR T-cell therapy in children and young adults with B-ALL. J. Clin. Oncol. 39, 1650–1659 (2021).
Pulsipher, M. A. 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. 3, 66–81 (2022).
Wayne, A. S. et al. Three-year results from phase I of ZUMA-4: KTE-X19 in pediatric relapsed/refractory acute lymphoblastic leukemia. Haematologica 108, 747–760 (2023).
Ghorashian, S. et al. Enhanced CAR T cell expansion and prolonged persistence in pediatric patients with ALL treated with a low-affinity CD19 CAR. Nat. Med. 25, 1408–1414 (2019).
Roddie, C. et al. Durable responses and low toxicity after fast off-rate CD19 chimeric antigen receptor-T therapy in adults with relapsed or refractory B-cell acute lymphoblastic leukemia. J. Clin. Oncol. 39, 3352–3363 (2021).
Hines, M. R. et al. Immune effector cell-associated hemophagocytic lymphohistiocytosis-like syndrome. Transpl. Cell Ther. 29, 438.e1–438.e16 (2023).
Lee, D. W. et al. ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol. Blood Marrow Transpl. 25, 625–638 (2019).
Rouce, R. H. et al. Evolution of tisagenlecleucel use for the treatment of pediatric and young adult relapsed/refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL): Center for International Blood & Marrow Transplant Research (CIBMTR) registry results [abstract]. J. Clin. Oncol. 42, 10016 (2024).
Maude, S. L. et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 371, 1507–1517 (2014).
Teachey, D. T. et al. Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discov. 6, 664–679 (2016).
Gardner, R. A. et al. Preemptive mitigation of CD19 CAR T-cell cytokine release syndrome without attenuation of antileukemic efficacy. Blood 134, 2149–2158 (2019).
Kadauke, S. 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. 39, 920–930 (2021).
Hogan, L. E. et al. Severe toxicity and poor efficacy of reinduction chemotherapy are associated with overall poor outcomes in relapsed B-cell acute lymphoblastic leukemia: a report from the Children’s Oncology Group AALL1331 trial. Haematologica 110, 2930–2941 (2025).
Oskarsson, T. et al. Treatment-related mortality in relapsed childhood acute lymphoblastic leukemia. Pediatr. Blood Cancer 65, e26909 (2018).
Peters, C. et al. Total body irradiation or chemotherapy conditioning in childhood ALL: a multinational, randomized, noninferiority phase III study. J. Clin. Oncol. 39, 295–307 (2021).
Naik, S. et al. Outcomes after second hematopoietic stem cell transplantations in pediatric patients with relapsed hematological malignancies. Biol. Blood Marrow Transpl. 21, 1266–1272 (2015).
Kamal, M. et al. Patient-reported outcomes for cancer patients with hematological malignancies undergoing chimeric antigen receptor T cell therapy: a systematic review. Transpl. Cell Ther. 27, 390.e1–390.e7 (2021).
Bansal, M., Sharma, K. K., Vatsa, M. & Bakhshi, S. Comparison of health-related quality of life of children during maintenance therapy with acute lymphoblastic leukemia versus siblings and healthy children in India. Leuk. Lymphoma 54, 1036–1041 (2013).
Fardell, J. E. et al. Health-related quality of life of children on treatment for acute lymphoblastic leukemia: a systematic review. Pediatr. Blood Cancer 64, e26489 (2017).
Bhakta, N. et al. The cumulative burden of surviving childhood cancer: an initial report from the St Jude Lifetime Cohort Study (SJLIFE). Lancet 390, 2569–2582 (2017).
Dixon, S. B. et al. Specific causes of excess late mortality and association with modifiable risk factors among survivors of childhood cancer: a report from the Childhood Cancer Survivor Study cohort. Lancet 401, 1447–1457 (2023).
Eichinger, A. et al. Incidence of subsequent malignancies after total body irradiation-based allogeneic HSCT in children with ALL — long-term follow-up from the prospective ALL-SCT 2003 trial. Leukemia 36, 2567–2576 (2022).
Shalabi, H. et al. Beyond the storm — subacute toxicities and late effects in children receiving CAR T cells. Nat. Rev. Clin. Oncol. 18, 363–378 (2021).
Kampouri, E., Walti, C. S., Gauthier, J. & Hill, J. A. Managing hypogammaglobulinemia in patients treated with CAR-T-cell therapy: key points for clinicians. Expert Rev. Hematol. 15, 305–320 (2022).
Epperly, R. & Shah, N. N. Long-term follow-up of CD19-CAR T-cell therapy in children and young adults with B-ALL. Hematol. Am. Soc. Hematol Educ. Program. 2023, 77–83 (2023).
Fried, S. et al. Early and late hematologic toxicity following CD19 CAR-T cells. Bone Marrow Transpl. 54, 1643–1650 (2019).
Nair, M. S. et al. Development of ALL-Hematotox (ALL-HT): predicting post CAR T-cell hematotoxicity in B-cell acute lymphoblastic leukemia. Blood 145, 1136–1148 (2025).
Levine, J. E. et al. Pooled safety analysis of tisagenlecleucel in children and young adults with B cell acute lymphoblastic leukemia. J. Immunother. Cancer 9, e002287 (2021).
Naik, S. et al. Characterization and prediction of prolonged severe neutropenia in pediatric patients receiving tisagenlecleucel. Blood Adv. 9, 5070–5084 (2025).
Hsieh, E. M. et al. Low rate of subsequent malignant neoplasms after CD19 CAR T-cell therapy. Blood Adv. 6, 5222–5226 (2022).
Tix, T. et al. Second primary malignancies after CAR T-Cell therapy: a systematic review and meta-analysis of 5,517 lymphoma and myeloma patients. Clin. Cancer Res. 30, 4690–4700 (2024).
US Food and Drug Administration. FDA investigating serious risk of T-cell malignancy following BCMA-directed or CD19-directed autologous chimeric antigen receptor (CAR) T cell immunotherapies. fda.gov https://www.fda.gov/vaccines-blood-biologics/safety-availability-biologics/fda-investigating-serious-risk-t-cell-malignancy-following-bcma-directed-or-cd19-directed-autologous (2023).
Dulery, R. et al. T cell malignancies after CAR T cell therapy in the DESCAR-T registry. Nat. Med. 31, 1130–1133 (2025).
Lamble, A. J. et al. Risk of T-cell malignancy after CAR T-cell therapy in children, adolescents, and young adults. Blood Adv. 8, 3544–3548 (2024).
Bhojwani, D. & Pui, C. H. Relapsed childhood acute lymphoblastic leukaemia. Lancet Oncol. 14, e205–e217 (2013).
Rheingold, S. R. et al. Determinants of survival after first relapse of acute lymphoblastic leukemia: a Children’s Oncology Group study. Leukemia 38, 2382–2394 (2024).
Tallen, G. et al. Long-term outcome in children with relapsed acute lymphoblastic leukemia after time-point and site-of-relapse stratification and intensified short-course multidrug chemotherapy: results of trial ALL-REZ BFM 90. J. Clin. Oncol. 28, 2339–2347 (2010).
Leahy, A. B. et al. Impact of high-risk cytogenetics on outcomes for children and young adults receiving CD19-directed CAR T-cell therapy. Blood 139, 2173–2185 (2022).
Lamble, A. J. et al. Preinfusion factors impacting relapse immunophenotype following CD19 CAR T cells. Blood Adv. 7, 575–585 (2023).
Ghorashian, S. 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. 9, e766–e775 (2022).
Pan, J. et al. Frequent occurrence of CD19-negative relapse after CD19 CAR T and consolidation therapy in 14 TP53-mutated r/r B-ALL children. Leukemia 34, 3382–3387 (2020).
Cox, W. P. J. et al. Loss of p53 impairs death receptor expression and confers resistance to CD19 CAR T-cell therapy in BCP-ALL. Blood Neoplasia 2, 100060 (2025).
Weber, E. W. et al. Transient rest restores functionality in exhausted CAR-T cells through epigenetic remodeling. Science 372, eaba1786 (2021).
Mavuluri, J. et al. GPR65 inactivation in tumor cells drives antigen-independent CAR-T cell resistance via macrophage remodeling. Cancer Discov. 15, 1018–1036 (2025).
Masih, K. E. et al. A stem cell epigenome is associated with primary nonresponse to CD19 CAR T cells in pediatric acute lymphoblastic leukemia. Blood Adv. 7, 4218–4232 (2023).
Singh, N. et al. Impaired death receptor signaling in leukemia causes antigen-independent resistance by inducing CAR T-cell dysfunction. Cancer Discov. 10, 552–567 (2020).
Barsan, V. et al. Tisagenlecleucel utilisation and outcomes across refractory, first relapse and multiply relapsed B-cell acute lymphoblastic leukemia: a retrospective analysis of real-world patterns. EClinicalMedicine 65, 102268 (2023).
Bader, P. et al. CD19 CAR T cells are an effective therapy for posttransplant relapse in patients with B-lineage ALL: real-world data from Germany. Blood Adv. 7, 2436–2448 (2023).
Maude, S. L., Barrett, D., Teachey, D. T. & Grupp, S. A. Managing cytokine release syndrome associated with novel T cell-engaging therapies. Cancer J. 20, 119–122 (2014).
Myers, R. M. et al. Blinatumomab nonresponse and high-disease burden are associated with inferior outcomes after CD19-CAR for B-ALL. J. Clin. Oncol. 40, 932–944 (2022).
Dourthe, M. E. et al. Determinants of CD19-positive vs CD19-negative relapse after tisagenlecleucel for B-cell acute lymphoblastic leukemia. Leukemia 35, 3383–3393 (2021).
Leahy, A. B. 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. 8, e711–e722 (2021).
Fabrizio, V. A. et al. Tisagenlecleucel outcomes in relapsed/refractory extramedullary ALL: a Pediatric Real World CAR Consortium report. Blood Adv. 6, 600–610 (2022).
Ortiz-Maldonado, V. et al. Results of ARI-0001 CART19 cell therapy in patients with relapsed/refractory CD19-positive acute lymphoblastic leukemia with isolated extramedullary disease. Am. J. Hematol. 97, 731–739 (2022).
Bachy, E. et al. A real-world comparison of tisagenlecleucel and axicabtagene ciloleucel CAR T cells in relapsed or refractory diffuse large B cell lymphoma. Nat. Med. 28, 2145–2154 (2022).
Jacoby, E. et al. CD19 CAR T-cells for pediatric relapsed acute lymphoblastic leukemia with active CNS involvement: a retrospective international study. Leukemia 36, 1525–1532 (2022).
Holland, E. M. et al. Characterization of extramedullary disease in B-ALL and response to CAR T-cell therapy. Blood Adv. 6, 2167–2182 (2022).
Shahid, S. 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. Transpl. Cell Ther. 28, 72.e1–72.e8 (2022).
Brivio, E. et al. A phase 1 study of inotuzumab ozogamicin in pediatric relapsed/refractory acute lymphoblastic leukemia (ITCC-059 study). Blood 137, 1582–1590 (2021).
Brown, P. A. 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 325, 833–842 (2021).
Pillai, V. et al. CAR T-cell therapy is effective for CD19-dim B-lymphoblastic leukemia but is impacted by prior blinatumomab therapy. Blood Adv. 3, 3539–3549 (2019).
Sahai, I. et al. Incidence of preexisting B-cell aplasia in B-ALL: implications for post-CAR T-cell monitoring. Blood Adv. 8, 6329–6333 (2024).
Gardner, R. et al. Acquisition of a CD19-negative myeloid phenotype allows immune escape of MLL-rearranged B-ALL from CD19 CAR-T-cell therapy. Blood 127, 2406–2410 (2016).
Gupta, S. et al. Blinatumomab in standard-risk B-cell acute lymphoblastic leukemia in children. N. Engl. J. Med. 392, 875–891 (2025).
Ceolin, V. et al. Outcome of chimeric antigen receptor T-cell therapy following treatment with inotuzumab ozogamicin in children with relapsed or refractory acute lymphoblastic leukemia. Leukemia 37, 53–60 (2023).
Dekker, L. et al. Fludarabine exposure predicts outcome after CD19 CAR T-cell therapy in children and young adults with acute leukemia. Blood Adv. 6, 1969–1976 (2022).
Fabrizio, V. A. et al. Optimal fludarabine lymphodepletion is associated with improved outcomes after CAR T-cell therapy. Blood Adv. 6, 1961–1968 (2022).
Ramos, C. A. et al. Anti-CD30 CAR-T cell therapy in relapsed and refractory Hodgkin lymphoma. J. Clin. Oncol. 38, 3794–3804 (2020).
Hirayama, A. V. et al. The response to lymphodepletion impacts PFS in patients with aggressive non-Hodgkin lymphoma treated with CD19 CAR T cells. Blood 133, 1876–1887 (2019).
Klebanoff, C. A., Khong, H. T., Antony, P. A., Palmer, D. C. & Restifo, N. P. Sinks, suppressors and antigen presenters: how lymphodepletion enhances T cell-mediated tumor immunotherapy. Trends Immunol. 26, 111–117 (2005).
Turtle, C. J. et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J. Clin. Invest. 126, 2123–2138 (2016).
Scordo, M. et al. Identifying an optimal fludarabine exposure for improved outcomes after axi-cel therapy for aggressive B-cell non-Hodgkin lymphoma. Blood Adv. 7, 5579–5585 (2023).
von Stackelberg, A. et al. Phase I/phase II study of blinatumomab in pediatric patients with relapsed/refractory acute lymphoblastic leukemia. J. Clin. Oncol. 34, 4381–4389 (2016).
Locatelli, F. et al. Blinatumomab in pediatric relapsed/refractory B-cell acute lymphoblastic leukemia: RIALTO expanded access study final analysis. Blood Adv. 6, 1004–1014 (2022).
Gore, L. et al. Survival after blinatumomab treatment in pediatric patients with relapsed/refractory B-cell precursor acute lymphoblastic leukemia. Blood Cancer J. 8, 80 (2018).
Llaurador, G. et al. Blinatumomab therapy is associated with favorable outcomes after allogeneic hematopoietic cell transplantation in pediatric patients with B cell acute lymphoblastic leukemia. Transpl. Cell Ther. 30, 217–227 (2024).
Gokbuget, N. et al. Blinatumomab for minimal residual disease in adults with B-cell precursor acute lymphoblastic leukemia. Blood 131, 1522–1531 (2018).
Litzow, M. R. et al. Blinatumomab for MRD-negative acute lymphoblastic leukemia in adults. N. Engl. J. Med. 391, 320–333 (2024).
van der Sluis, I. M. et al. Blinatumomab added to chemotherapy in infant lymphoblastic leukemia. N. Engl. J. Med. 388, 1572–1581 (2023).
Aldoss, I. et al. Correlates of resistance and relapse during blinatumomab therapy for relapsed/refractory acute lymphoblastic leukemia. Am. J. Hematol. 92, 858–865 (2017).
Slaney, C. Y., Wang, P., Darcy, P. K. & Kershaw, M. H. CARs versus BiTEs: a comparison between T cell-redirection strategies for cancer treatment. Cancer Discov. 8, 924–934 (2018).
Coyle, L. et al. Open-label, phase 2 study of blinatumomab as second salvage therapy in adults with relapsed/refractory aggressive B-cell non-Hodgkin lymphoma. Leuk. Lymphoma 61, 2103–2112 (2020).
Dufner, V. et al. Long-term outcome of patients with relapsed/refractory B-cell non-Hodgkin lymphoma treated with blinatumomab. Blood Adv. 3, 2491–2498 (2019).
Zugmaier, G., Subklewe, M. & Locatelli, F. Immunotherapy with blinatumomab in B-cell acute lymphoblastic leukemia: a narrative review of efficacy, toxicity, and patient management in relapse and consolidation. Ann. Blood https://doi.org/10.21037/aob-25-5 (2025).
Jabbour, E. et al. Single agent subcutaneous blinatumomab for advanced acute lymphoblastic leukemia. Am. J. Hematol. 99, 586–595 (2024).
O’Brien, M. M. 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. 40, 956–967 (2022).
Kantarjian, H. M. et al. Inotuzumab ozogamicin for relapsed/refractory acute lymphoblastic leukemia in the INO-VATE trial: CD22 pharmacodynamics, efficacy, and safety by baseline CD22. Clin. Cancer Res. 27, 2742–2754 (2021).
Pennesi, 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 36, 1516–1524 (2022).
Bhojwani, D. et al. Inotuzumab ozogamicin in pediatric patients with relapsed/refractory acute lymphoblastic leukemia. Leukemia 33, 884–892 (2019).
Kantarjian, H. M. et al. Inotuzumab ozogamicin versus standard therapy for acute lymphoblastic leukemia. N. Engl. J. Med. 375, 740–753 (2016).
Kouhi, A. et al. Brain disposition of antibody-based therapeutics: dogma, approaches and perspectives. Int. J. Mol. Sci. 22, 6442 (2021).
DeAngelo, D. J. et al. Inotuzumab ozogamicin for relapsed/refractory acute lymphoblastic leukemia: outcomes by disease burden. Blood Cancer J. 10, 81 (2020).
Pennesi, E. et al. Inotuzumab ozogamicin combined with chemotherapy in pediatric B-cell precursor CD22+ acute lymphoblastic leukemia: results of the phase IB ITCC-059 trial. Haematologica 109, 3157–3166 (2024).
Kayser, S. et al. Impact of inotuzumab ozogamicin on outcome in relapsed or refractory acute B-cell lymphoblastic leukemia patients prior to allogeneic hematopoietic stem cell transplantation and risk of sinusoidal obstruction syndrome/venous occlusive disease. Haematologica 109, 1385–1392 (2024).
Rubinstein, J. D. & O’Brien, M. M. Inotuzumab ozogamicin in B-cell precursor acute lymphoblastic leukemia: efficacy, toxicity, and practical considerations. Front. Immunol. 14, 1237738 (2023).
Grupp, S. A. et al. Defibrotide plus best standard of care compared with best standard of care alone for the prevention of sinusoidal obstruction syndrome (HARMONY): a randomised, multicentre, phase 3 trial. Lancet Haematol. 10, e333–e345 (2023).
Kebriaei, P. et al. Management of important adverse events associated with inotuzumab ozogamicin: expert panel review. Bone Marrow Transpl. 53, 449–456 (2018).
Brivio, E. et al. Sinusoidal obstruction syndrome in children with CD22+ B-cell precursors acute lymphoblastic leukemia (BCP-ALL) treated with inotuzumab ozogamicin in trial ITCC-059: risk factors and outcomes [abstract]. Blood 144, 310–311 (2024).
Pulsipher, M. A. et al. IgH-V(D)J NGS-MRD measurement pre- and early post-allotransplant defines very low- and very high-risk ALL patients. Blood 125, 3501–3508 (2015).
Abdel-Azim, H. et al. Next generation sequence minimal residual disease (NGS-MRD) predicts outstanding event free survival (EFS) regardless of hematopoietic cell transplantation (HCT) preparative approach or graft alpha/beta depletion in children with acute lymphoblastic leukemia (ALL) [abstract]. Blood 134, 4624 (2019).
Gaballa, M. R. et al. Blinatumomab maintenance after allogeneic hematopoietic cell transplantation for B-lineage acute lymphoblastic leukemia. Blood 139, 1908–1919 (2022).
Summers, C. et al. Hematopoietic cell transplantation after CD19 chimeric antigen receptor T cell-induced acute lymphoblastic lymphoma remission confers a leukemia-free survival advantage. Transpl. Cell Ther. 28, 21–29 (2022).
Fabrizio, V. A. et al. Low toxicity and favorable overall survival in relapsed/refractory B-ALL following CAR T cells and CD34-selected T-cell depleted allogeneic hematopoietic cell transplant. Bone Marrow Transpl. 55, 2160–2169 (2020).
Contreras, C. F. et al. Clinical utilization of blinatumomab and inotuzumab immunotherapy in children with relapsed or refractory B-acute lymphoblastic leukemia. Pediatr. Blood Cancer 68, e28718 (2021).
Gibson, A. et al. Combination low-intensity chemotherapy plus inotuzumab ozogamicin, blinatumomab and rituximab for pediatric patients with relapsed/refractory B-cell acute lymphoblastic leukemia. Haematologica 109, 3042–3047 (2024).
Kantarjian, H. et al. Results of salvage therapy with mini-hyper-CVD and inotuzumab ozogamicin with or without blinatumomab in pre-B acute lymphoblastic leukemia. J. Hematol. Oncol. 16, 44 (2023).
Jabbour, E. et al. Chemoimmunotherapy with inotuzumab ozogamicin combined with mini-hyper-CVD, with or without blinatumomab, is highly effective in patients with Philadelphia chromosome-negative acute lymphoblastic leukemia in first salvage. Cancer 124, 4044–4055 (2018).
Kantarjian, H. et al. Hyper-CVAD and sequential blinatumomab without and with inotuzumab in young adults with newly diagnosed Philadelphia chromosome-negative B-cell acute lymphoblastic leukemia. Am. J. Hematol. 100, 402–407 (2025).
Cordoba, S. et al. CAR T cells with dual targeting of CD19 and CD22 in pediatric and young adult patients with relapsed or refractory B cell acute lymphoblastic leukemia: a phase 1 trial. Nat. Med. 27, 1797–1805 (2021).
Shalabi, H. et al. CD19/22 CAR T cells in children and young adults with B-ALL: phase 1 results and development of a novel bicistronic CAR. Blood 140, 451–463 (2022).
Spiegel, J. Y. et al. CAR T cells with dual targeting of CD19 and CD22 in adult patients with recurrent or refractory B cell malignancies: a phase 1 trial. Nat. Med. 27, 1419–1431 (2021).
Pan, J. et al. Sequential CD19-22 CAR T therapy induces sustained remission in children with r/r B-ALL. Blood 135, 387–391 (2020).
Wang, T. et al. Coadministration of CD19- and CD22-directed chimeric antigen receptor T-cell therapy in childhood B-cell acute lymphoblastic leukemia: a single-arm, multicenter, phase II trial. J. Clin. Oncol. 41, 1670–1683 (2023).
Hunger, S. P. et al. Improved survival for children and adolescents with acute lymphoblastic leukemia between 1990 and 2005: a report from the Children’s Oncology Group. J. Clin. Oncol. 30, 1663–1669 (2012).
Turcotte, L. M. et al. Real-world cost of pediatric acute lymphoblastic leukemia care among commercially insured individuals in the United States: effect of era and age at diagnosis. JCO Oncol. Pract. 18, e1750–e1761 (2022).
Sarkar, R. R., Gloude, N. J., Schiff, D. & Murphy, J. D. Cost-effectiveness of chimeric antigen receptor T-cell therapy in pediatric relapsed/refractory B-cell acute lymphoblastic leukemia. J. Natl Cancer Inst. 111, 719–726 (2019).
Laetsch, T. et al. Evolving evidence-based value assessment of one-time therapies: tisagenlecleucel as a case study. Appl. Health Econ. Health Policy 22, 749–765 (2024).
Palani, H. K. et al. Safety, efficacy and total cost of point-of-care manufactured anti-CD19 CAR-T cell therapy in India: VELCART trial. Mol. Ther. Oncol. 33, 200977 (2025).
Rehman, M. A. & Arshad, H. A discount on the cost of cancer: India’s homegrown CAR-T cell therapy. Blood Cell Ther. 7, 121–123 (2024).
Martinez-Cibrian, N. et al. The academic point-of-care anti-CD19 chimeric antigen receptor T-cell product varnimcabtagene autoleucel (ARI-0001 cells) shows efficacy and safety in the treatment of relapsed/refractory B-cell non-Hodgkin lymphoma. Br. J. Haematol. 204, 525–533 (2024).
Goncalves, E. CAR-T cell therapies: patient access and affordability solutions. Future Sci. OA 11, 2483613 (2025).
Cliff, E. R. S. et al. High cost of chimeric antigen receptor T-cells: challenges and solutions. Am. Soc. Clin. Oncol. Educ. Book. 43, e397912 (2023).
Geethakumari, P. R., Ramasamy, D. P., Dholaria, B., Berdeja, J. & Kansagra, A. Balancing quality, cost, and access during delivery of newer cellular and immunotherapy treatments. Curr. Hematol. Malig. Rep. 16, 345–356 (2021).
Gupta, S., Maude, S. L., O’Brien, M. M., Rau, R. E. & McNeer, J. L. How the COG is approaching the high-risk patient with ALL: incorporation of immunotherapy into frontline treatment. Clin. Lymphoma Myeloma Leuk. 20, S8–S11 (2020).
Bartram, J., Ancliff, P. & Vora, A. How I treat infant acute lymphoblastic leukemia. Blood 145, 35–42 (2025).
Pieters, R. et al. Outcome of infants younger than 1 year with acute lymphoblastic leukemia treated with the Interfant-06 protocol: results from an international phase III randomized study. J. Clin. Oncol. 37, 2246–2256 (2019).
Brown, P., Pieters, R. & Biondi, A. How I treat infant leukemia. Blood 133, 205–214 (2019).
Fong, D., Tiwari, R., Acker, C., Clough, L. & Willert, J. Leukapheresis and tisagenlecleucel manufacturing outcomes in patients age <3 years with relapsed/refractory acute lymphoblastic leukemia. Transpl. Cell Ther. 29, 579.e1–579.e10 (2023).
Annesley, C. et al. Feasibility and favorable responses following investigational CAR T-cell therapy for relapsed and refractory infant ALL. Blood Adv. 9, 2068–2078 (2025).
Rabin, K. R. et al. Outcomes in children, adolescents, and young adults with Down syndrome and all: a report from the Children’s Oncology Group. J. Clin. Oncol. 42, 218–227 (2024).
Laetsch, T. W. et al. Tisagenlecleucel in pediatric and young adult patients with Down syndrome-associated relapsed/refractory acute lymphoblastic leukemia. Leukemia 36, 1508–1515 (2022).
Aldoss, I. et al. Donor-derived CD19-targeted chimeric antigen receptor T cells in adult transplant recipients with relapsed/refractory acute lymphoblastic leukemia. Blood Cancer J. 13, 107 (2023).
Del Bufalo, F. et al. Allogeneic, donor-derived, second-generation, CD19-directed CAR-T cells for the treatment of pediatric relapsed/refractory BCP-ALL. Blood 142, 146–157 (2023).
Dourthe, M. E. et al. Success of donor-derived CAR-T cells after failure of autologous CD19 CAR-T cells (tisagenlecleucel) in B-cell acute lymphoblastic leukaemia. Br. J. Haematol. 205, 373–377 (2024).
Laetsch, T. W., DuBois, S. G., Bender, J. G., Macy, M. E. & Moreno, L. Opportunities and challenges in drug development for pediatric cancers. Cancer Discov. 11, 545–559 (2021).
Singh, K. et al. Breaking the silence: challenges and opportunities in pediatric drug development. Pediatr. Res. 98, 807–812 (2025).
Martinez-Gamboa, D. A. et al. CAR T-cell therapy landscape in pediatric, adolescent and young adult oncology — a comprehensive analysis of clinical trials. Crit. Rev. Oncol. Hematol. 209, 104648 (2025).
Pearson, A. D. et al. Paediatric Strategy Forum for medicinal product development of chimeric antigen receptor T-cells in children and adolescents with cancer: ACCELERATE in collaboration with the European Medicines Agency with participation of the Food and Drug Administration. Eur. J. Cancer 160, 112–133 (2022).
Locke, F. L. et al. Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): a single-arm, multicentre, phase 1-2 trial. Lancet Oncol. 20, 31–42 (2019).
Locke, F. L. et al. Axicabtagene ciloleucel as second-line therapy for large B-cell lymphoma. N. Engl. J. Med. 386, 640–654 (2022).
Neelapu, S. S. et al. Axicabtagene ciloleucel car T-cell therapy in refractory large B-cell lymphoma. N. Engl. J. Med. 377, 2531–2544 (2017).
Schuster, S. J. et al. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N. Engl. J. Med. 380, 45–56 (2019).
Minard-Colin, V. et al. Rituximab for high-risk, mature B-cell non-Hodgkin’s lymphoma in children. N. Engl. J. Med. 382, 2207–2219 (2020).
Minard-Colin, V. et al. Efficacy and safety of tisagenlecleucel in pediatric and young adult patients (pts) with relapsed or refractory (r/r) mature B-cell non-Hodgkin lymphoma (NHL): the phase II BIANCA study [abstract S255]. HemaSphere 6, 156–157 (2022).
Liu, Y. et al. Sequential different B-cell antigen-targeted CAR T-cell therapy for pediatric refractory/relapsed Burkitt lymphoma. Blood Adv. 6, 717–730 (2022).
Bender, J. D. et al. Real-world use of tisagenlecleucel in children and young adults with relapsed or refractory B-cell lymphomas. Blood Adv. 8, 4164–4168 (2024).
Minson, A. G. & Dickinson, M. J. New bispecific antibodies in diffuse large B-cell lymphoma. Haematologica 110, 1483–1499 (2025).
Schipani, M. et al. Bispecific monoclonal antibodies in diffuse large B-cell lymphoma: dawn of a new era in targeted therapy. Cancers 17, 3258 (2025).
Shah, N. N. 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. 38, 1938–1950 (2020).
Xiao, X., Ho, M., Zhu, Z., Pastan, I. & Dimitrov, D. S. Identification and characterization of fully human anti-CD22 monoclonal antibodies. MAbs 1, 297–303 (2009).
CARGO Therapeutics. CARGO Therapeutics to discontinue FIRCE-1 phase 2 study of firi-cel; advances remaining programs while evaluating strategic options. GlobeNewswire https://www.globenewswire.com/news-release/2025/01/29/3017565/0/en/CARGO-Therapeutics-to-Discontinue-FIRCE-1-Phase-2-Study-of-Firi-cel-Advances-Remaining-Programs-While-Evaluating-Strategic-Options.html (2025).
Ghorashian, S. et al. CD19/CD22 targeting with cotransduced CAR T cells to prevent antigen-negative relapse after CAR T-cell therapy for B-cell ALL. Blood 143, 118–123 (2024).
Dai, H. et al. Bispecific CAR-T cells targeting both CD19 and CD22 for therapy of adults with relapsed or refractory B cell acute lymphoblastic leukemia. J. Hematol. Oncol. 13, 30 (2020).
Yang, T. et al. Prominent efficacy and good safety of sequential CD19 and CD22 CAR-T therapy in relapsed/refractory adult B-cell acute lymphoblastic leukemia. Exp. Hematol. Oncol. 14, 2 (2025).
Frey, N. V. et al. CART22-65s co-administered with huCART19 in adult patients with relapsed or refractory ALL [abstract]. Blood 138, 469 (2021).
Marcus, A., Waks, T. & Eshhar, Z. Redirected tumor-specific allogeneic T cells for universal treatment of cancer. Blood 118, 975–983 (2011).
Kochenderfer, J. N. et al. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood 122, 4129–4139 (2013).
Benjamin, R. et al. Genome-edited, donor-derived allogeneic anti-CD19 chimeric antigen receptor T cells in paediatric and adult B-cell acute lymphoblastic leukaemia: results of two phase 1 studies. Lancet 396, 1885–1894 (2020).
Benjamin, R. et al. UCART19, a first-in-class allogeneic anti-CD19 chimeric antigen receptor T-cell therapy for adults with relapsed or refractory B-cell acute lymphoblastic leukaemia (CALM): a phase 1, dose-escalation trial. Lancet Haematol. 9, e833–e843 (2022).
Ottaviano, G. et al. Phase 1 clinical trial of CRISPR-engineered CAR19 universal T cells for treatment of children with refractory B cell leukemia. Sci. Transl. Med. 14, eabq3010 (2022).
Qasim, W. et al. Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells. Sci. Transl. Med. 9, eaaj2013 (2017).
Xiao, X. et al. CD19-CAR-DNT cells (RJMty19) in patients with relapsed or refractory large B-cell lymphoma: a phase 1, first-in-human study. EClinicalMedicine 70, 102516 (2024).
Pan, J. et al. Allogeneic CD5-specific CAR-T therapy for relapsed/refractory T-ALL: a phase 1 trial. Nat. Med. 31, 126–136 (2025).
Chiesa, R. et al. Base-edited CAR7 T cells for relapsed T-cell acute lymphoblastic leukemia. N. Engl. J. Med. 389, 899–910 (2023).
Ghobadi, A. et al. Phase 1/2 trial of anti-CD7 allogeneic WU-CART-007 for patients with relapsed/refractory T-cell malignancies. Blood 146, 1163–1173 (2025).
Li, S. et al. Eradication of T-ALL Cells by CD7-targeted Universal CAR-T cells and initial test of ruxolitinib-based CRS management. Clin. Cancer Res. 27, 1242–1246 (2021).
Xu, X. et al. HLA fully-mismatched sibling-derived CD7 CAR-T therapy bridging to haploidentical hematopoietic stem cell transplantation for hepatosplenic γδ T-cell lymphoma. Cell Transpl. 32, 9636897231194265 (2023).
Zhang, Y. et al. Allogenic and autologous anti-CD7 CAR-T cell therapies in relapsed or refractory T-cell malignancies. Blood Cancer J. 13, 61 (2023).
Pemmaraju, N. et al. CD123-directed allogeneic chimeric-antigen receptor T-cell therapy (CAR-T) in blastic plasmacytoid dendritic cell neoplasm (BPDCN): clinicopathological insights. Leuk. Res. 121, 106928 (2022).
Brown, C. E. et al. Off-the-shelf, steroid-resistant, IL13Rα2-specific CAR T cells for treatment of glioblastoma. Neuro Oncol. 24, 1318–1330 (2022).
Hill, L. C. et al. Antitumor efficacy and safety of unedited autologous CD5.CAR T cells in relapsed/refractory mature T-cell lymphomas. Blood 143, 1231–1241 (2024).
Hu, Y. et al. Genetically modified CD7-targeting allogeneic CAR-T cell therapy with enhanced efficacy for relapsed/refractory CD7-positive hematological malignancies: a phase I clinical study. Cell Res. 32, 995–1007 (2022).
Lu, P. et al. Naturally selected CD7 CAR-T therapy without genetic manipulations for T-ALL/LBL: first-in-human phase 1 clinical trial. Blood 140, 321–334 (2022).
Zhang, M. et al. Autologous nanobody-derived fratricide-resistant CD7-CAR T-cell therapy for patients with relapsed and refractory T-cell acute lymphoblastic leukemia/lymphoma. Clin. Cancer Res. 28, 2830–2843 (2022).
Oh, B. L. Z. et al. Fratricide-resistant CD7-CAR T cells in T-ALL. Nat. Med. 30, 3687–3696 (2024).
Mu, W. et al. Case report: differential diagnosis of highly amplified anti-CD5 CAR T cells and relapsed lymphoma cells in a patient with refractory ALK positive anaplastic large cell lymphoma. Front. Immunol. 14, 1280007 (2023).
Shah, N. N. et al. CD33 CAR T-Cells (CD33CART) for children and young adults with relapsed/refractory AML: dose-escalation results from a phase I/II multicenter trial [abstract]. Blood 142, 771 (2023).
Naik, S. et al. Safety and anti-leukemic activity of CD123-CAR T cells in pediatric patients with AML: preliminary results from a phase 1 trial [abstract]. Blood 140, 4584–4585 (2022).
Naik, S. et al. CD123-CAR T cells manufactured in the presence of dasatinib induce high grade CRS and/or IEC-HS without improving efficacy in pediatric patients with recurrent/refractory leukemia [abstract]. Blood 144, 2076 (2024).
Geyer, M. B. et al. CD371-targeted CAR T cells secreting interleukin-18 exhibit robust expansion and clear refractory acute myeloid leukemia. Blood 146, 3163–3174 (2025).
Bhagwat, A. S. et al. Cytokine-mediated CAR T therapy resistance in AML. Nat. Med. 30, 3697–3708 (2024).
Haubner, S. et al. Cooperative CAR targeting to selectively eliminate AML and minimize escape. Cancer Cell 41, 1871–1891.e6 (2023).
Caruso, S. et al. Safe and effective off-the-shelf immunotherapy based on CAR.CD123-NK cells for the treatment of acute myeloid leukaemia. J. Hematol. Oncol. 15, 163 (2022).
Perkins, S. M., Shinohara, E. T., DeWees, T. & Frangoul, H. Outcome for children with metastatic solid tumors over the last four decades. PLoS ONE 9, e100396 (2014).
Pule, M. A. et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat. Med. 14, 1264–1270 (2008).
Louis, C. U. et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood 118, 6050–6056 (2011).
Li, C. H. et al. Long-term outcomes of GD2-directed CAR-T cell therapy in patients with neuroblastoma. Nat. Med. 31, 1125–1129 (2025).
Albelda, S. M. CAR T cell therapy for patients with solid tumours: key lessons to learn and unlearn. Nat. Rev. Clin. Oncol. 21, 47–66 (2024).
Ligon, J. A. et al. INSPIRED Symposium Part 4B: chimeric antigen receptor T cell correlative studies — established findings and future priorities. Transpl. Cell Ther. 30, 155–170 (2024).
Ramakrishna, S., Barsan, V. & Mackall, C. Prospects and challenges for use of CAR T cell therapies in solid tumors. Expert Opin. Biol. Ther. 20, 503–516 (2020).
Dyson, K. A. et al. Emerging trends in immunotherapy for pediatric sarcomas. J. Hematol. Oncol. 12, 78 (2019).
Theruvath, J. et al. Locoregionally administered B7-H3-targeted CAR T cells for treatment of atypical teratoid/rhabdoid tumors. Nat. Med. 26, 712–719 (2020).
Monje, M. et al. Intravenous and intracranial GD2-CAR T cells for H3K27M+ diffuse midline gliomas. Nature 637, 708–715 (2025).
Dominguez-Prieto, V. et al. Understanding CAR T cell therapy and its role in ovarian cancer and peritoneal carcinomatosis from ovarian cancer. Front. Oncol. 13, 1104547 (2023).
Kaczanowska, S. et al. Immune determinants of CAR-T cell expansion in solid tumor patients receiving GD2 CAR-T cell therapy. Cancer Cell 42, 35–51 e8 (2024).
Labanieh, L. & Mackall, C. L. CAR immune cells: design principles, resistance and the next generation. Nature 614, 635–648 (2023).
Heitzeneder, S. et al. GPC2-CAR T cells tuned for low antigen density mediate potent activity against neuroblastoma without toxicity. Cancer Cell 40, 53–69.e9 (2022).
Bosse, K. R. et al. Identification of GPC2 as an oncoprotein and candidate immunotherapeutic target in high-risk neuroblastoma. Cancer Cell 32, 295–309.e12 (2017).
Yarmarkovich, M. et al. Targeting of intracellular oncoproteins with peptide-centric CARs. Nature 623, 820–827 (2023).
Majzner, R. G. et al. GD2-CAR T cell therapy for H3K27M-mutated diffuse midline gliomas. Nature 603, 934–941 (2022).
Heczey, A. et al. CAR T cells administered in combination with lymphodepletion and PD-1 inhibition to patients with neuroblastoma. Mol. Ther. 25, 2214–2224 (2017).
Ye, Z. et al. Phenotypic plasticity of myeloid cells in glioblastoma development, progression, and therapeutics. Oncogene 40, 6059–6070 (2021).
Tcyganov, E., Mastio, J., Chen, E. & Gabrilovich, D. I. Plasticity of myeloid-derived suppressor cells in cancer. Curr. Opin. Immunol. 51, 76–82 (2018).
Del Bufalo, F. et al. GD2-CART01 for relapsed or refractory high-risk neuroblastoma. N. Engl. J. Med. 388, 1284–1295 (2023).
Steffin, D. et al. Interleukin-15-armoured GPC3 CAR T cells for patients with solid cancers. Nature 637, 940–946 (2025).
Jansen, L., Wienke, J., Molkenbur, R., Rossig, C. & Meissner, R. B7-H3 in the tumor microenvironment: implications for CAR T cell therapy in pediatric solid tumors. Cancer Metastasis Rev. 44, 77 (2025).
Giudice, A. M. et al. Reprogramming the neuroblastoma tumor immune microenvironment to enhance GPC2 CAR T cells. Mol. Ther. 33, 4552–4569 (2025).
Nguyen, D. T. et al. CAR T cell locomotion in solid tumor microenvironment. Cells 11, 1974 (2022).
Hegde, M. et al. Autologous HER2-specific CAR T cells after lymphodepletion for advanced sarcoma: a phase 1 trial. Nat. Cancer 5, 880–894 (2024).
Pinto, N. et al. STRIvE-02: a first-in-human phase I study of systemically administered B7-H3 chimeric antigen receptor T cells for patients with relapsed/refractory solid tumors. J. Clin. Oncol. 42, 4163–4172 (2024).
Vitanza, N. A. et al. Intracerebroventricular B7-H3-targeting CAR T cells for diffuse intrinsic pontine glioma: a phase 1 trial. Nat. Med. 31, 861–868 (2025).
Schett, G. & June, C. H. CAR T cells in autoimmune disease: on the road to remission. Immunity 57, 2705–2709 (2024).
Schett, G. et al. Advancements and challenges in CAR T cell therapy in autoimmune diseases. Nat. Rev. Rheumatol. 20, 531–544 (2024).
Chung, J. B., Brudno, J. N., Borie, D. & Kochenderfer, J. N. Chimeric antigen receptor T cell therapy for autoimmune disease. Nat. Rev. Immunol. 24, 830–845 (2024).
Auth, J. et al. CD19-targeting CAR T-cell therapy in patients with diffuse systemic sclerosis: a case series. Lancet Rheumatol. 7, e83–e93 (2025).
Haghikia, A. et al. Anti-CD19 CAR T cells for refractory myasthenia gravis. Lancet Neurol. 22, 1104–1105 (2023).
Muller, F. et al. CD19 CAR T-cell therapy in autoimmune disease — a case series with follow-up. N. Engl. J. Med. 390, 687–700 (2024).
Wobma, H. et al. CAR T cell therapy for children with rheumatic disease: the time is now. Nat. Rev. Rheumatol. 21, 494–506 (2025).
Mikhael, J., Fowler, J. & Shah, N. Chimeric antigen receptor T-cell therapies: barriers and solutions to access. JCO Oncol. Pract. 18, 800–807 (2022).
Pearson, A. D. J. et al. New models for the development of and access to CAR T-cell therapies for children and adolescents with cancer: an ACCELERATE multistakeholder analysis. Lancet Oncol. 26, e214–e224 (2025).
Hall, A. G. et al. Access to chimeric antigen receptor T cell clinical trials in underrepresented populations: a multicenter cohort study of pediatric and young adult acute lymphobastic leukemia patients. Transpl. Cell Ther. 29, 356.e1–356.e7 (2023).
Gardner, R. A. et al. ACT to sustain: adoptive cell therapy to sustain access to non-commercialized genetically modified cell therapies. Transpl. Cell Ther. 30, 776–787 (2024).
Rossig, C. et al. Chimeric antigen receptor (CAR) T-cell products for pediatric cancers: why alternative development paths are needed. J. Clin. Oncol. 42, 253–257 (2024).
Mackall, C. L. et al. Enhancing pediatric access to cell and gene therapies. Nat. Med. 30, 1836–1846 (2024).
Park, J. H. et al. CD19 CAR T-cell therapy and prophylactic anakinra in relapsed or refractory lymphoma: phase 2 trial interim results. Nat. Med. 29, 1710–1717 (2023).
Frigault, M. J. et al. A phase II trial of anakinra for the prevention of CAR-T cell mediated neurotoxicity [abstract]. Blood 138, 2814 (2021).
Strati, P. et al. A phase 1 study of prophylactic anakinra to mitigate ICANS in patients with large B-cell lymphoma. Blood Adv. 7, 6785–6789 (2023).
Diorio, C. et al. Anakinra utilization in refractory pediatric CAR T-cell associated toxicities. Blood Adv. 6, 3398–3403 (2022).
Shah, N. N., Johnson, B. D., Fenske, T. S., Raj, R. V. & Hari, P. Intrathecal chemotherapy for management of steroid-refractory CAR T-cell-associated neurotoxicity syndrome. Blood Adv. 4, 2119–2122 (2020).
Rejeski, K. et al. Immune effector cell-associated hematotoxicity: EHA/EBMT consensus grading and best practice recommendations. Blood 142, 865–877 (2023).
Rejeski, K., Jain, M. D., Shah, N. N., Perales, M. A. & Subklewe, M. Immune effector cell-associated haematotoxicity after CAR T-cell therapy: from mechanism to management. Lancet Haematol. 11, e459–e470 (2024).
Jain, T., Olson, T. S. & Locke, F. L. How I treat cytopenias after CAR T-cell therapy. Blood 141, 2460–2469 (2023).
Rejeski, K. et al. Safety and feasibility of stem cell boost as a salvage therapy for severe hematotoxicity after CD19 CAR T-cell therapy. Blood Adv. 6, 4719–4725 (2022).
Lipsitt, A. et al. Allogeneic CD34+ selected hematopoietic stem cell boost following CAR T-cell therapy in a patient with prolonged cytopenia and active infection. Pediatr. Blood Cancer 70, e30166 (2023).
Mahdi, J. et al. Tumor inflammation-associated neurotoxicity. Nat. Med. 29, 803–810 (2023).
Borowitz, M. J. et al. Prognostic significance of minimal residual disease in high risk B-ALL: a report from Children’s Oncology Group Study AALL0232. Blood 126, 964–971 (2015).
Minson, K. A. et al. t(17;19) in children with acute lymphocytic leukemia: a report of 3 cases and a review of the literature. Case Rep. Hematol. 2013, 563291 (2013).
Hogan, L. E. et al. Children’s Oncology Group AALL1331: phase III trial of blinatumomab in children, adolescents, and young adults with low-risk B-cell all in first relapse. J. Clin. Oncol. 41, 4118–4129 (2023).
Hastings, C. et al. Late isolated central nervous system relapse in childhood B-cell acute lymphoblastic leukemia treated with intensified systemic therapy and delayed reduced dose cranial radiation: a report from the Children’s Oncology Group Study AALL02P2. Pediatr. Blood Cancer 68, e29256 (2021).
Moskop, A. et al. Real-world use of tisagenlecleucel in infant acute lymphoblastic leukemia. Blood Adv. 6, 4251–4255 (2022).
Lust, H. et al. Real world outcomes for AYA patients with relapsed/refractory B-cell ALL receiving CD19-directed CAR T-cell therapy. Transplant. Cell. Ther. 30, S39–S40 (2024).
Pan, J. et al. Donor-derived CD7 chimeric antigen receptor T cells for T-cell acute lymphoblastic leukemia: first-in-human, phase I trial. J. Clin. Oncol. 39, 3340–3351 (2021).
Sallman, D. A. et al. Ameli-01: a phase I trial of UCART123v1. 2, an anti-CD123 allogeneic CAR-T cell product, in adult patients with relapsed or refractory (R/R) CD123+ acute myeloid leukemia (AML). Blood 140, 2371–2373 (2022).
Ehninger, G. et al. Phase 1 dose escalation study of the rapidly switchable universal CAR-T therapy unicar-T-CD123 in relapsed/refractory AML [abstract]. Blood 140, 2367–2368 (2022).
Wermke, M. et al. Updated results from a phase I dose escalation study of the rapidly-switchable universal CAR-T therapy uniCAR-T-CD123 in relapsed/refractory AML [abstract]. Blood 142, 3465 (2023).
Tambaro, F. P. et al. Autologous CD33-CAR-T cells for treatment of relapsed/refractory acute myelogenous leukemia. Leukemia 35, 3282–3286 (2021).
Wang, Q. S. et al. Treatment of CD33-directed chimeric antigen receptor-modified T cells in one patient with relapsed and refractory acute myeloid leukemia. Mol. Ther. 23, 184–191 (2015).
Sallman, D. A. et al. Phase 1/1b safety study of PRGN-3006 ultracar-T in patients with relapsed or refractory CD33-positive acute myeloid leukemia and higher risk myelodysplastic syndromes. Blood 140, 10313–10315 (2022).
Appelbaum, J. et al. Drug-regulated CD33-targeted CAR T cells control AML using clinically optimized rapamycin dosing. J. Clin. Invest. 134, e162593 (2024).
Zhang, H. et al. Characteristics of anti-CLL1 based CAR-T therapy for children with relapsed or refractory acute myeloid leukemia: the multi-center efficacy and safety interim analysis. Leukemia 36, 2596–2604 (2022).
Zhang, H. et al. Anti-CLL1 chimeric antigen receptor T-cell therapy in children with relapsed/refractory acute myeloid leukemia. Clin. Cancer Res. 27, 3549–3555 (2021).
Pei, K. et al. Anti-CLL1-based CAR T-cells with 4-1-BB or CD28/CD27 stimulatory domains in treating childhood refractory/relapsed acute myeloid leukemia. Cancer Med. 12, 9655–9661 (2023).
Jin, X. et al. First-in-human phase I study of CLL-1 CAR-T cells in adults with relapsed/refractory acute myeloid leukemia. J. Hematol. Oncol. 15, 88 (2022).
Zhang, X. et al. A phase I clinical trial of CLL-1 CAR-T cells for the treatment of relapsed/refractory acute myeloid leukemia in adults [abstract]. Blood 142, 2106 (2023).
Ma, Y. J. et al. Successful application of PD-1 knockdown CLL-1 CAR-T therapy in two AML patients with post-transplant relapse and failure of anti-CD38 CAR-T cell treatment. Am. J. Cancer Res. 12, 615–621 (2022).
Liu, F. et al. First-in-human CLL1-CD33 compound CAR T cell therapy induces complete remission in patients with refractory acute myeloid leukemia: update on phase 1 clinical trial [abstract]. Blood 132, 901 (2018).
Baumeister, S. H. et al. Phase I trial of autologous CAR T cells targeting NKG2D ligands in patients with AML/MDS and multiple myeloma. Cancer Immunol. Res. 7, 100–112 (2019).
Sallman, D. A. et al. CYAD-01, an autologous NKG2D-based CAR T-cell therapy, in relapsed or refractory acute myeloid leukaemia and myelodysplastic syndromes or multiple myeloma (THINK): haematological cohorts of the dose escalation segment of a phase 1 trial. Lancet Haematol. 10, e191–e202 (2023).
Pinto N. et al. First-in-human comparison of second-versus third-generation L1CAM-specific CAR T cells in patients with primary refractory or relapsed neuroblastoma. Preprint at Research Square https://doi.org/10.21203/rs.3.rs-3859120/v1 (2024).
Vitanza, N. A. et al. Intraventricular B7-H3 CAR T cells for diffuse intrinsic pontine glioma: preliminary first-in-human bioactivity and safety. Cancer Discov. 13, 114–131 (2023).
Heczey, A. et al. Anti-GD2 CAR-NKT cells in relapsed or refractory neuroblastoma: updated phase 1 trial interim results. Nat. Med. 29, 1379–1388 (2023).
Straathof, K. et al. Antitumor activity without on-target off-tumor toxicity of GD2-chimeric antigen receptor T cells in patients with neuroblastoma. Sci. Transl. Med. 12, eabd6169 (2020).
Yu, L. et al. GD2-specific chimeric antigen receptor-modified T cells for the treatment of refractory and/or recurrent neuroblastoma in pediatric patients. J. Cancer Res. Clin. Oncol. 148, 2643–2652 (2022).
Liu, Z. et al. Safety and antitumor activity of GD2-specific 4SCAR-T cells in patients with glioblastoma. Mol. Cancer 22, 3 (2023).
Ahmed, N. et al. HER2-specific chimeric antigen receptor-modified virus-specific T cells for progressive glioblastoma: a phase 1 dose-escalation trial. JAMA Oncol. 3, 1094–1101 (2017).
Donovan, L. K. et al. Locoregional delivery of CAR T cells to the cerebrospinal fluid for treatment of metastatic medulloblastoma and ependymoma. Nat. Med. 26, 720–731 (2020).
Brown, C. E. et al. Regression of glioblastoma after chimeric antigen receptor T-cell therapy. N. Engl. J. Med. 375, 2561–2569 (2016).
Brown, C. E. et al. Locoregional delivery of IL-13Rα2-targeting CAR-T cells in recurrent high-grade glioma: a phase 1 trial. Nat. Med. 30, 1001–1012 (2024).
O’Rourke, D. M. et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci. Transl. Med. 9, eaaa0984 (2017).
Goff, S. L. et al. Pilot trial of adoptive transfer of chimeric antigen receptor-transduced T cells targeting EGFRvIII in patients with glioblastoma. J. Immunother. 42, 126–135 (2019).
Wang, C. M. et al. Autologous T cells expressing CD30 chimeric antigen receptors for relapsed or refractory Hodgkin lymphoma: an open-label phase I trial. Clin. Cancer Res. 23, 1156–1166 (2017).
Ahmed, S. et al. Updated results and correlative analysis: autologous CD30. CAR-T-cell therapy in patients with relapsed or refractory classical hodgkin lymphoma (CHARIOT trial) [abstract]. Blood 140, 7496–7497 (2022).
Fu, Q. et al. RUNX-3-expressing CAR T cells targeting glypican-3 in patients with heavily pretreated advanced hepatocellular carcinoma: a phase I trial. EClinicalMedicine 63, 102175 (2023).
Shi, D. et al. Chimeric antigen receptor-glypican-3 T-cell therapy for advanced hepatocellular carcinoma: results of phase I trials. Clin. Cancer Res. 26, 3979–3989 (2020).
Acknowledgements
The work of N.N.S is supported in part by the Intramural Research Program, Center of Cancer Research, National Cancer Institute and National Institutes of Health (NIH) Clinical Center (grant ZIA BC 011823). S.L.M. is a Scholar in Clinical Research of The Leukaemia & Lymphoma Society.
Author information
Authors and Affiliations
Contributions
L.S., K.McN., A.J.L., F.G.C., A.B., F.C., K.J.C., L.G., M.L., S.L.M., M.A.P., S.R., C.S., C.M.C., D.B., R.A.G., S.G. and N.N.S. researched data for the article. L.S., K.McN., A.J.L., F.G.C., A.B., F.C., K.J.C., L.G., M.L., S.L.M., M.A.P., S.R., S.R.R., S.K.S., A.S., C.S., C.M.C., D.B., R.A.G., S.G. and N.N.S. wrote the manuscript. All authors contributed substantially to discussions of the content and reviewed and/or edited the manuscript before submission. This Review was conceptualized and led by L.S. and N.N.S.
Corresponding authors
Ethics declarations
Competing interests
L.G. has received travel funding and honoraria from Amgen. S.L.M. has received research support for clinical trials sponsored by Luminary Therapeutics. M.A.P. has served on advisory boards for Autolus, bluebird bio, Cargo Therapeutics, Garuda/Stratus Therapeutics, GentiBio, Mesoblast, Novartis and Pfizer; and had received research support from Adaptive Biotech and Miltenyi Biotec. M.Q. has received honoraria from Medexus. S.R.R. declares consultancy roles for Amgen and Pfizer. C.R. has received honoraria from Amgen and Novartis. C.M.C. reports honoraria from Bayer, Nektar Therapeutics and Novartis; and has equity interest in Elephas. D.B. declares research funding from Wugen. R.A.G. has patents related to CAR T cell technologies licensed with royalties paid from Juno Therapeutics. N.N.S. declares research funding from Cargo Therapeutics, Lentigen and Vor Bio; has attended advisory board meetings (no honoraria) for ImmunoACT, Sobi and Vor Bio; and receives royalties from Cargo Therapeutics. The other authors declare no competing interests.
Peer review
Peer review information
Nature Reviews Clinical Oncology thanks E. Jacoby, M. Maus and the other, anonymous, reviewers for their contribution to the peer review of this work.
Additional information
Disclaimer
The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products, or organizations imply endorsement by the U.S. Government.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary information
Rights and permissions
About this article
Cite this article
Schultz, L., McNerney, K., Lamble, A.J. et al. The quintessential role for CAR T cell therapy in children, adolescents and young adults with cancer. Nat Rev Clin Oncol (2026). https://doi.org/10.1038/s41571-025-01115-w
Accepted:
Published:
Version of record:
DOI: https://doi.org/10.1038/s41571-025-01115-w


