Zum Inhalt
Erschienen in:

Open Access 07.05.2025 | short review

Blinatumomab is changing the standard of care paradigms of newly diagnosed and relapsed pediatric B-cell acute lymphoblastic leukemia

verfasst von: Prof. MD Andishe Attarbaschi, Fiona Poyer

Erschienen in: memo - Magazine of European Medical Oncology | Ausgabe 2/2025

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Summary

Since 2016, a relevant number of phase I, II, and III trials performed with children and adolescents with CD19-positive B‑cell acute lymphoblastic leukemia (B-ALL) have studied the use of the bi-specific T‑cell engager antibody blinatumomab with respect to both improving disease outcomes and reducing therapy-related toxicities. All studies reported to date, from infants to older adults with B‑ALL, have shown a definitive advantage for blinatumomab intercalated into multidrug chemotherapy regimens or, in some trials, as a replacement of intensive chemotherapy phases. Conclusively, blinatumomab-based therapy is rapidly being established as the new standard of care for treatment of pediatric patients ≥ 1 month of age with CD19 + B-ALL as part of consolidation of a multiphase chemotherapy. The story of the success of blinatumomab in childhood B‑ALL is summarized in the current review article.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

While acute lymphoblastic leukemia (ALL) is the most common malignant disease of childhood and adolescence, accounting for 25–30% of all cancers, the first relapse of ALL represents the sixth most common pediatric cancer and remains the leading cause of pediatric cancer-related mortality. Newly diagnosed ALL is curable with frontline therapy in 80–85% of children and adolescence, and the first relapse of ALL can be cured with salvage therapy in 50–60% of patients [1, 2]. However, it must be taken into account that the results achieved to date with intensive chemotherapy are associated with severe acute toxicities, long treatment duration, and also late effects. Further intensification of conventional chemotherapies (and allogeneic hematopoietic stem cell transplantation [allo-HSCT]) is not feasible and, in particular, not justifiable [3]. The success of allo-HSCT has already impressively demonstrated the efficacy of immunotherapy for childhood ALL (graft-vs.-leukemia effect; [4]).
It is thus expected that new antibody-based or cellular immunotherapies will significantly change the treatment spectrum of children and adolescents with ALL in the coming years, aiming not only to enhance outcomes, but also to mitigate treatment toxicity [5]. CD19 and CD22 are highly expressed on B‑ALL blasts and CD38 and CD7 on T‑ALL blasts, and these antigens are currently of great interest for targeted immunotherapeutic interventions. The successful use of monoclonal antibodies (mABs), unconjugated (i.e., daratumumab, anti-CD38), conjugated to toxins (i.e., inotuzumab ozogamicin, anti-CD22 + calicheamicin), or as bi-specific antibodies (i.e., blinatumomab, anti-CD19/CD3), has already been impressively shown in the relapsed and refractory (r/r) setting of pediatric ALL [510]. “Off-the-shelf” mABs and antibody–drug conjugates have the advantage of being tumor-specific rather than patient-specific and can, therefore, be produced industrially, stocked easily, and used immediately in patients without waiting times. However, they are generally not curative as monotherapies, but “only” induce and/or maintain complete morphological/molecular remission, which needs to be consolidated by allo-HSCT (or chimeric antigen receptor [CAR] T cells) in the r/r setting of ALL [11]. By contrast, adding mAB to a multiphase chemotherapy protocol in the context of the frontline ALL setting usually aims at improving disease-free survival (DFS) and/or reducing acute toxicities for all risk groups without the need for allo-HSCT. The concept of combining the MHC-independent antigen recognition of mAB and the effector function of T cells was a breakthrough in the field of immunotherapy of malignant diseases and led to the development and broad clinical testing of blinatumomab but also, of course, of CAR T cells (i.e., tisagenlecleucel).

Blinatumomab in pediatric B-ALL

Blinatumomab is currently administered in children with B‑ALL via continuous intravenous (IV) infusion for 28 days per cycle and targets CD19 on B cells and CD3 on T cells, thereby allowing T cells to recognize and destroy CD19-positive leukemia cells [12]. The first phase I/II trial (NCT01471782) and the successive phase II trial (RIALTO, NCT02187354) tested single-agent blinatumomab for r/r pediatric B‑ALL (refractory disease, ≥  2nd relapsed B‑ALL, any relapse after allo-HSCT; [1315]). Together they demonstrated (1) its anti-leukemic efficacy in morphologically visible disease (≥ 25% blasts), with 52% of the responders (39%) achieving minimal residual disease (MRD) negativity; (2) its enhanced anti-leukemic activity in minimal residual bone marrow (BM) disease (52% achieved remission with MRD response); (3) its favorable toxicity profile; and (4) its anti-leukemic activity across all age and risk groups. Neurological toxicities and cytokine release syndrome were the most relevant but well manageable toxicities reported. The successive NEUF study, involving a real-world pediatric cohort of r/r B‑ALL and providing blinatumomab in an expanded access program, also showed that blinatumomab is effective, with > 50% of patients with relapse achieving remission and those treated in an MRD-positive setting also doing extremely well [16].
In addition to the RIALTO trial, three randomized phase III trials of the International Relapsed B‑ALL (high-risk relapse, NCT02393859) consortium and the Children’s Oncology Group AALL1331 (COG, low-, intermediate-, and high-risk relapse; NCT02101853, NCT02101853) for a first relapse of B‑ALL were conducted [1719]. The improved outcome measures (event- and disease-free survival, overall survival, MRD response, toxicity, and realization of allo-HSCT) obtained with blinatumomab in the context of multiphase therapy protocols established blinatumomab as a new standard of care for post-reinduction consolidation in pediatric patients with a high- and intermediate-risk first relapse of B‑ALL and with a standard-risk first relapse of B‑ALL with BM involvement [1719]. Notably, similarly dismal outcomes for isolated extramedullary relapses were reported with both standard of care chemotherapy and blinatumomab-based therapy in AALL1731 [17], leading to the pursuit of alternative approaches in this relapse setting.
A recent pilot trial also added one cycle of blinatumomab to post-induction Interfant-06 consolidation therapy for infants with newly diagnosed KMT2A-rearranged B‑ALL (EudraCT number 2016-004674-17; [20]). The addition of blinatumomab to this chemotherapy regimen was feasible, it markedly increased the rate of MRD-negative remission, and, most importantly, it led to significantly improved clinical outcomes with 2‑year DFS of 81.6% and overall survival (OS) of 93.3% compared to historic Interfant-06 chemotherapy-only outcomes of 49.4% and 65.8%, respectively [21]. Consequently, this major breakthrough established blinatumomab as a new standard of care in the context of a multiphase therapy of newly diagnosed infant KMT2A-rearranged B‑ALL. The addition of two cycles of post-induction blinatumomab with some chemotherapy reduction is now under prospective evaluation in a larger cohort of infants with newly diagnosed KMT2A-rearranged B‑ALL in the current international Interfant-21 trial (NCT05327894) with the goal of validating the superior outcomes observed in the Interfant-06/blinatumomab pilot trial and reducing chemotherapy-associated toxicity.
The AIEOP-BFM ALL 2017 trial (NCT03643276) was the first randomized phase III trial to evaluate a multi-agent chemotherapy regimen without or with the addition of blinatumomab in children with newly diagnosed intermediate-risk B‑ALL patients. This study also randomized patients with high-risk B‑ALL to two cycles of blinatumomab versus two intensive consolidation blocks with the goal of understanding the potential for improved DFS and OS and for decreased infectious and other acute toxicity [22]. Although outcome data are still pending, the toxicity profile appeared to be significantly in favor of blinatumomab with no life-threatening events recorded and the majority of adverse events of special interest in the blinatumomab arm to be neurological disorders (mainly grade 2 or 3 seizures; [22]). If upcoming analyses of outcome data show at least non-inferiority in terms of anti-leukemia efficacy, replacement of the intensive chemotherapy blocks by blinatumomab will become the new standard of care for consolidation treatment of newly diagnosed non-infant patients with high-risk B‑ALL.
The COG also performed a recent phase III trial (NCT03914625) of children with newly diagnosed standard-risk B‑ALL who had an average or higher risk of relapse [23]. Patients were randomized to receive standard-of-care chemotherapy alone or chemotherapy with the addition of two non-sequential cycles of blinatumomab. The randomization was permanently stopped following an interim analysis demonstrating clearly superior results for the blinatumomab arm compared to the chemotherapy-only arm, with 3‑year DFS for those with standard average-risk B‑ALL of 97.5 ± 1.3% vs. 90.2 ± 2.3% and for those with standard high-risk B‑ALL of 94.1 ± 2.5% vs. 84.8 ± 3.8% [24]. Notably, the incidence of non-fatal sepsis/catheter-related infections increased in children with standard average-risk B‑ALL treated in the blinatumomab arm, not just during blinatumomab cycles with continuous IV access, but well into maintenance therapy for reasons not yet fully understood [24]. Nevertheless, the addition of blinatumomab to therapy was established as the new standard of care for consolidation treatment of newly diagnosed non-infant patients with standard-risk B‑ALL.
Blinatumomab has also been documented as an effective consolidation approach for pediatric and adolescent patients with B‑ALL with chemotherapy intolerance during frontline treatment due to severe acute toxicities (e.g., infections, typhlitis, or L‑asparaginase-associated acute pancreatitis). The United Kingdom ALL group studied a cohort of 105 pediatric patients who received one to two cycles of blinatumomab as replacement for post-remission consolidation chemotherapy, 82% of whom were treated due to chemotherapy intolerance and 18% due to persistent leukemia/MRD positivity after induction therapy [25]. Blinatumomab was well tolerated and very effective, with 97% of patients achieving MRD-negative complete remission (CR). Event-free survival rates were similar to a matched chemotherapy patient cohort [25]. In line with the preliminary results of the AIEOP-BFM ALL 2017 trial, this study suggested that blinatumomab could replace intensive chemotherapy blocks in a high-risk B‑ALL population.
Collectively, according to recent data clearly demonstrating the superiority of blinatumomab-based therapy in infants with KMT2A-rearranged B‑ALL and children with SR B‑ALL and the similarly impressive rates of MRD-negative CR of adult patients with Philadelphia-negative B‑ALL treated with post-induction blinatumomab, the US Food and Drug Administration approved blinatumomab as consolidation therapy for patients \(\geq\)28 days of age with newly diagnosed B‑ALL. It is anticipated the European Medicines Agency will similarly update approved indications for blinatumomab in the near future. As a consequence of this success story, blinatumomab will be further investigated in soon-to-open and planned frontline pediatric B‑ALL trials, including for patients with Philadelphia-positive and ABL class Ph-like B‑ALL in combination with tyrosine kinase inhibitors via the EsPhALL2022/COG AALL2131 pilot study (NCT06124157), and in those with first relapsed B‑ALL treated at early time points of protocol therapy (now also replacing very early consolidation chemotherapy elements; [2729]). Although generally less toxic than intensive cytotoxic chemotherapy and amenable to outpatient administration, blinatumomab has potential for specific immunologic and neurologic adverse events (e.g., cytokine release syndrome, seizure, encephalopathy), and its current need for continuous infusion and IV access may be logistically challenging [30]. A recent subcutaneous formulation of blinatumomab has been tested in adults with r/r B‑ALL and demonstrated high CR rates and impressive rates of MRD clearance, representing an emerging viable alternative to continuous IV delivery and meriting pediatric-specific investigation [31].
In conclusion, the use of blinatumomab now gives children and adolescents with resistant or multiple relapsed B‑ALL the chance of achieving molecular remissions and definitive cures through therapy with successive allo-HSCT or CAR T cells. However, as blinatumomab also improved outcomes in the frontline and first-relapse setting of pediatric B‑ALL, but mostly when “only” given on top of a multiphase chemotherapy protocol, its use at earlier time points of our study protocols seems warranted in order to replace conventional chemotherapy. Thereby, acute toxicities could be further reduced and with improved early response to therapy, high-risk group allocations and allo-HSCT rates could be decreased.
Take-home message
Blinatumomab intercalation into therapy for patients with relapsed and now newly diagnosed B‑ALL across the age spectrum has remarkably improved clinical outcomes. Additional pediatric-specific clinical trial investigation of blinatumomab-based therapies with therapy reduction where feasible is warranted.

Acknowledgements

We thank Dr. Sarah K. Tasian for a fruitful and inspiring discussion of the content of the present review.

Conflict of interest

A. Attarbaschi: Honoraria: Jazz Pharmaceuticals, Amgen, and Novartis; Consulting or Advisory Role: Jazz Pharmaceuticals, Amgen, and Novartis; Travel, Accommodations, Expenses: Jazz Pharmaceuticals. F. Poyer declares that she has no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

Abo für kostenpflichtige Inhalte

Literatur
1.
Zurück zum Zitat Conter V, Bartram CR, Valsecchi MG, et al. Molecular response to treatment redefines all prognostic factors in children and adolescents with B‑cell precursor acute lymphoblastic leukemia: results in 3184 patients of the AIEOP-BFM ALL 2000 study. Blood. 2010;115(16):3206–14.CrossRefPubMed Conter V, Bartram CR, Valsecchi MG, et al. Molecular response to treatment redefines all prognostic factors in children and adolescents with B‑cell precursor acute lymphoblastic leukemia: results in 3184 patients of the AIEOP-BFM ALL 2000 study. Blood. 2010;115(16):3206–14.CrossRefPubMed
2.
Zurück zum Zitat Rizzari C, Moricke A, Valsecchi MG, et al. Incidence and Characteristics of Hypersensitivity Reactions to PEG-asparaginase Observed in 6136 Children With Acute Lymphoblastic Leukemia Enrolled in the AIEOP-BFM ALL 2009 Study Protocol. Hemasphere. 2023;7(6):e893.CrossRefPubMedPubMedCentral Rizzari C, Moricke A, Valsecchi MG, et al. Incidence and Characteristics of Hypersensitivity Reactions to PEG-asparaginase Observed in 6136 Children With Acute Lymphoblastic Leukemia Enrolled in the AIEOP-BFM ALL 2009 Study Protocol. Hemasphere. 2023;7(6):e893.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Peters C, Dalle JH, Locatelli F, et al. Total body irradiation or chemotherapy conditioning in childhood ALL: a multinational, randomized, noninferiority phase III study. J Clin Oncol. 2021;39(4):295–307.CrossRefPubMed Peters C, Dalle JH, Locatelli F, et al. Total body irradiation or chemotherapy conditioning in childhood ALL: a multinational, randomized, noninferiority phase III study. J Clin Oncol. 2021;39(4):295–307.CrossRefPubMed
5.
Zurück zum Zitat Brivio E, Bautista F, Zwaan CM. Naked antibodies and antibody-drug conjugates: targeted therapy for childhood acute lymphoblastic leukemia. Haematologica. 2024;109(6):1700–12.CrossRefPubMedPubMedCentral Brivio E, Bautista F, Zwaan CM. Naked antibodies and antibody-drug conjugates: targeted therapy for childhood acute lymphoblastic leukemia. Haematologica. 2024;109(6):1700–12.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Gore L, Locatelli F, Zugmaier G, et al. Survival after blinatumomab treatment in pediatric patients with relapsed/refractory B‑cell precursor acute lymphoblastic leukemia. Blood Cancer J. 2018;8(9):80.CrossRefPubMedPubMedCentral Gore L, Locatelli F, Zugmaier G, et al. Survival after blinatumomab treatment in pediatric patients with relapsed/refractory B‑cell precursor acute lymphoblastic leukemia. Blood Cancer J. 2018;8(9):80.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Handgretinger R, Zugmaier G, Henze G, Kreyenberg H, Lang P, von Stackelberg A. Complete remission after blinatumomab-induced donor T‑cell activation in three pediatric patients with post-transplant relapsed acute lymphoblastic leukemia. Leukemia. 2011;25(1):181–4.CrossRefPubMed Handgretinger R, Zugmaier G, Henze G, Kreyenberg H, Lang P, von Stackelberg A. Complete remission after blinatumomab-induced donor T‑cell activation in three pediatric patients with post-transplant relapsed acute lymphoblastic leukemia. Leukemia. 2011;25(1):181–4.CrossRefPubMed
8.
Zurück zum Zitat Pennesi E, Brivio E, Ammerlaan ACJ, 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. 2024;109(10):3157–66.CrossRefPubMedPubMedCentral Pennesi E, Brivio E, Ammerlaan ACJ, 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. 2024;109(10):3157–66.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Brivio E, Locatelli F, Lopez-Yurda M, et al. A phase 1 study of inotuzumab ozogamicin in pediatric relapsed/refractory acute lymphoblastic leukemia (ITCC-059 study). Blood. 2021;137(12):1582–90.CrossRefPubMedPubMedCentral Brivio E, Locatelli F, Lopez-Yurda M, et al. A phase 1 study of inotuzumab ozogamicin in pediatric relapsed/refractory acute lymphoblastic leukemia (ITCC-059 study). Blood. 2021;137(12):1582–90.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Bhatla T, Hogan LE, Teachey DT, et al. Daratumumab in pediatric relapsed/refractory acute lymphoblastic leukemia or lymphoblastic lymphoma: the DELPHINUS study. Blood. 2024;144(21):2237–47.CrossRefPubMed Bhatla T, Hogan LE, Teachey DT, et al. Daratumumab in pediatric relapsed/refractory acute lymphoblastic leukemia or lymphoblastic lymphoma: the DELPHINUS study. Blood. 2024;144(21):2237–47.CrossRefPubMed
11.
Zurück zum Zitat Buchmann S, Schrappe M, Baruchel A, et al. Remission, treatment failure, and relapse in pediatric ALL: an international consensus of the Ponte-di-Legno Consortium. Blood. 2022;139(12):1785–93.CrossRefPubMedPubMedCentral Buchmann S, Schrappe M, Baruchel A, et al. Remission, treatment failure, and relapse in pediatric ALL: an international consensus of the Ponte-di-Legno Consortium. Blood. 2022;139(12):1785–93.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Loffler A, Kufer P, Lutterbuse R, et al. A recombinant bispecific single-chain antibody, CD19 x CD3, induces rapid and high lymphoma-directed cytotoxicity by unstimulated T lymphocytes. Blood. 2000;95(6):2098–103.CrossRefPubMed Loffler A, Kufer P, Lutterbuse R, et al. A recombinant bispecific single-chain antibody, CD19 x CD3, induces rapid and high lymphoma-directed cytotoxicity by unstimulated T lymphocytes. Blood. 2000;95(6):2098–103.CrossRefPubMed
13.
Zurück zum Zitat Locatelli F, Zugmaier G, Mergen N, et al. Blinatumomab in pediatric patients with relapsed/refractory acute lymphoblastic leukemia: results of the RIALTO trial, an expanded access study. Blood Cancer J. 2020;10(7):77.CrossRefPubMedPubMedCentral Locatelli F, Zugmaier G, Mergen N, et al. Blinatumomab in pediatric patients with relapsed/refractory acute lymphoblastic leukemia: results of the RIALTO trial, an expanded access study. Blood Cancer J. 2020;10(7):77.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Locatelli F, Zugmaier G, Mergen N, et al. Blinatumomab in pediatric relapsed/refractory B‑cell acute lymphoblastic leukemia: RIALTO expanded access study final analysis. Blood Adv. 2022;6(3):1004–14.CrossRefPubMedPubMedCentral Locatelli F, Zugmaier G, Mergen N, et al. Blinatumomab in pediatric relapsed/refractory B‑cell acute lymphoblastic leukemia: RIALTO expanded access study final analysis. Blood Adv. 2022;6(3):1004–14.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat von Stackelberg A, Locatelli F, Zugmaier G, et al. Phase I/phase II study of blinatumomab in pediatric patients with relapsed/refractory acute lymphoblastic leukemia. J Clin Oncol. 2016;34(36):4381–9.CrossRef von Stackelberg A, Locatelli F, Zugmaier G, et al. Phase I/phase II study of blinatumomab in pediatric patients with relapsed/refractory acute lymphoblastic leukemia. J Clin Oncol. 2016;34(36):4381–9.CrossRef
16.
Zurück zum Zitat Locatelli F, Maschan A, Boissel N, et al. Pediatric patients with acute lymphoblastic leukemia treated with blinatumomab in a real-world setting: results from the NEUF study. Pediatr Blood Cancer. 2022;69(4):e29562.CrossRefPubMed Locatelli F, Maschan A, Boissel N, et al. Pediatric patients with acute lymphoblastic leukemia treated with blinatumomab in a real-world setting: results from the NEUF study. Pediatr Blood Cancer. 2022;69(4):e29562.CrossRefPubMed
17.
Zurück zum Zitat Brown PA, Ji L, Xu X, et al. A randomized phase 3 trial of Blinatumomab vs. chemotherapy as post-reinduction therapy in low risk (LR) first relapse of B‑acute lymphoblastic leukemia (B-ALL) in children and adolescents/young adults (AYas): a report from children’s oncology group study AALL1331. Blood. 2021;138(Supplement 1):363.CrossRef Brown PA, Ji L, Xu X, et al. A randomized phase 3 trial of Blinatumomab vs. chemotherapy as post-reinduction therapy in low risk (LR) first relapse of B‑acute lymphoblastic leukemia (B-ALL) in children and adolescents/young adults (AYas): a report from children’s oncology group study AALL1331. Blood. 2021;138(Supplement 1):363.CrossRef
18.
Zurück zum Zitat 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):833–42.CrossRefPubMedPubMedCentral 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):833–42.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat 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):843–54.CrossRefPubMedPubMedCentral 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):843–54.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat van der Sluis IM, de Lorenzo P, Kotecha RS, et al. Blinatumomab added to chemotherapy in infant lymphoblastic leukemia. N Engl J Med. 2023;388(17):1572–81.CrossRefPubMed van der Sluis IM, de Lorenzo P, Kotecha RS, et al. Blinatumomab added to chemotherapy in infant lymphoblastic leukemia. N Engl J Med. 2023;388(17):1572–81.CrossRefPubMed
21.
Zurück zum Zitat Pieters R, De Lorenzo P, Ancliffe P, 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. 2019;37(25):2246–56.CrossRefPubMed Pieters R, De Lorenzo P, Ancliffe P, 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. 2019;37(25):2246–56.CrossRefPubMed
22.
Zurück zum Zitat Schrappe M, Locatelli F, Valsecchi MG, et al. Pediatric patients with high-risk B‑cell ALL in first complete remission may benefit from less toxic immunotherapy with blinatumomab—results from randomized controlled phase 3 trial AIEOP-BFM ALL 2017. Blood. 2023;142:825–7.CrossRef Schrappe M, Locatelli F, Valsecchi MG, et al. Pediatric patients with high-risk B‑cell ALL in first complete remission may benefit from less toxic immunotherapy with blinatumomab—results from randomized controlled phase 3 trial AIEOP-BFM ALL 2017. Blood. 2023;142:825–7.CrossRef
23.
Zurück zum Zitat Conter V, Valsecchi MG, Parasole R, et al. Childhood high-risk acute lymphoblastic leukemia in first remission: results after chemotherapy or transplant from the AIEOP ALL 2000 study. Blood. 2014;123(10):1470–8.CrossRefPubMed Conter V, Valsecchi MG, Parasole R, et al. Childhood high-risk acute lymphoblastic leukemia in first remission: results after chemotherapy or transplant from the AIEOP ALL 2000 study. Blood. 2014;123(10):1470–8.CrossRefPubMed
24.
Zurück zum Zitat Gupta S, Rau RE, Kairalla JA, et al. Blinatumomab in standard-risk B‑cell acute lymphoblastic leukemia in children. N Engl J Med. 2025;392(9):875–91.CrossRefPubMed Gupta S, Rau RE, Kairalla JA, et al. Blinatumomab in standard-risk B‑cell acute lymphoblastic leukemia in children. N Engl J Med. 2025;392(9):875–91.CrossRefPubMed
25.
Zurück zum Zitat Hodder A, Mishra AK, Enshaei A, et al. Blinatumomab for first-line treatment of children and young persons with B‑ALL. J Clin Oncol. 2024;42(8):907–14.CrossRefPubMed Hodder A, Mishra AK, Enshaei A, et al. Blinatumomab for first-line treatment of children and young persons with B‑ALL. J Clin Oncol. 2024;42(8):907–14.CrossRefPubMed
26.
Zurück zum Zitat Litzow MR, Sun Z, Mattison RJ, et al. Blinatumomab for MRD-negative acute lymphoblastic leukemia in adults. N Engl J Med. 2024;391(4):320–33.CrossRefPubMedPubMedCentral Litzow MR, Sun Z, Mattison RJ, et al. Blinatumomab for MRD-negative acute lymphoblastic leukemia in adults. N Engl J Med. 2024;391(4):320–33.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Foa R, Bassan R, Vitale A, et al. Dasatinib-blinatumomab for Ph-positive acute lymphoblastic leukemia in adults. N Engl J Med. 2020;383(17):1613–23.CrossRefPubMed Foa R, Bassan R, Vitale A, et al. Dasatinib-blinatumomab for Ph-positive acute lymphoblastic leukemia in adults. N Engl J Med. 2020;383(17):1613–23.CrossRefPubMed
28.
Zurück zum Zitat Zeckanovic A, Mouttet B, Vinti L, et al. Update on long-term outcomes of a cohort of patients with TCF3::HLF positive acute lymphoblastic leukemia treated with blinatumomab and stem cell transplantation. Haematologica. 2025;. Zeckanovic A, Mouttet B, Vinti L, et al. Update on long-term outcomes of a cohort of patients with TCF3::HLF positive acute lymphoblastic leukemia treated with blinatumomab and stem cell transplantation. Haematologica. 2025;.
29.
Zurück zum Zitat Queudeville M, Schlegel P, Heinz AT, et al. Blinatumomab in pediatric patients with relapsed/refractory B‑cell precursor acute lymphoblastic leukemia. Eur J Haematol. 2021;106(4):473–83.CrossRefPubMed Queudeville M, Schlegel P, Heinz AT, et al. Blinatumomab in pediatric patients with relapsed/refractory B‑cell precursor acute lymphoblastic leukemia. Eur J Haematol. 2021;106(4):473–83.CrossRefPubMed
30.
Zurück zum Zitat Withycombe JS, Kubaney HR, Okada M, et al. Delivery of care for pediatric patients receiving blinatumomab: a children’s oncology group study. Cancer Nurs. 2024;47(6):451–9.CrossRefPubMed Withycombe JS, Kubaney HR, Okada M, et al. Delivery of care for pediatric patients receiving blinatumomab: a children’s oncology group study. Cancer Nurs. 2024;47(6):451–9.CrossRefPubMed
31.
Zurück zum Zitat Jabbour E, Zugmaier G, Agrawal V, et al. Single agent subcutaneous blinatumomab for advanced acute lymphoblastic leukemia. Am J Hematol. 2024;99(4):586–95.CrossRefPubMed Jabbour E, Zugmaier G, Agrawal V, et al. Single agent subcutaneous blinatumomab for advanced acute lymphoblastic leukemia. Am J Hematol. 2024;99(4):586–95.CrossRefPubMed
Metadaten
Titel
Blinatumomab is changing the standard of care paradigms of newly diagnosed and relapsed pediatric B-cell acute lymphoblastic leukemia
verfasst von
Prof. MD Andishe Attarbaschi
Fiona Poyer
Publikationsdatum
07.05.2025
Verlag
Springer Vienna
Erschienen in
memo - Magazine of European Medical Oncology / Ausgabe 2/2025
Print ISSN: 1865-5041
Elektronische ISSN: 1865-5076
DOI
https://doi.org/10.1007/s12254-025-01034-7