Elsevier

Leukemia Research

Volume 59, August 2017, Pages 136-141
Leukemia Research

Research paper
Haploidentical hematopoietic stem cell transplantation for pediatric Philadelphia chromosome-positive acute lymphoblastic leukemia in the imatinib era

https://doi.org/10.1016/j.leukres.2017.05.021Get rights and content

Highlights

  • Allo-HSCT remains an important curative option for children with Ph+ ALL.

  • The role of HID HSCT in pediatric patients with Ph+ ALL has not been reported.

  • The 5-year EFS and OS were 61.0% and 70.0% in HID HSCT for children with Ph+ ALL.

  • The 3-year incidence of relapse and non-relapse mortality were 22.7% and 16.4%.

  • BCR-ABL positivity on +30 day post-HSCT was a significant factor for relapse.

Abstract

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains an important curative option for children with Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) who have a poor response to chemotherapy plus imatinib. For such children, if there are no matched related or unrelated donors, alternative donor transplantation may be a choice. The role of haploidentical donor (HID) HSCT in pediatric patients with Ph+ ALL has not been reported. The study population included pediatric patients with Ph+ ALL who underwent HID-HSCT. BCR-ABL transcript levels were analyzed using real-time quantitative reverse transcription polymerase chain reaction. At a median follow-up of 34 months, the 5-year probabilities of event-free survival (EFS) and overall survival (OS) were 61.0% and 70.0%, respectively in HID HSCT. The 3-year incidence of relapse and non-relapse mortality was 22.7% and 16.4%. Multivariate analysis showed that the post-HSCT BCR-ABL transcript level on +30 day was a significant factor affecting relapse rate. HID HSCT for the treatment of pediatric patients with Ph+ ALL yielded promising long-term survival. Post-HSCT BCR-ABL transcript positivity was a significant factor for clinical relapse after allo-HSCT in the imatinib era.

Introduction

Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) in children was associated with a very poor prognosis despite the use of intensive chemotherapy, compared with Ph (−) ALL in children, before the advent of tyrosine kinase inhibitors (TKI). Allogeneic hematopoietic stem-cell transplantation (allo-HSCT) was the best curative therapy for children with Ph+ ALL [1], [2], [3]. Although chemotherapy combined with imatinib has greatly increased complete remission rate and remission duration in patients with Ph+ ALL, allo-HSCT is considered as an important curative treatment in younger adults with Ph+ ALL in many reports [4], [5], [6].

Recent studies showed that children and adolescents with Ph+ ALL treated with intensive chemotherapy plus intensive imatinib achieved an excellent long-term survival (70%), and allo-HSCT did not show an advantage [7]. In another key study (EsPhALL study), outcomes for pediatric patients with Ph+ ALL group who did not undergo HSCT appeared inferior to that observed in the transplanted cohort if they had available donors, but the number of patients who didn’t receive transplant was relative small [8]. Although allo-HSCT in first remission is not routinely recommended for all pediatric patients with Ph+ ALL, it may still be an important treatment option for children with Ph+ ALL, especially for those patients with poor response to chemotherapy plus TKI who have available donors.

Many studies showed that the addition of imatinib to chemotherapy in pediatric Ph+ ALL followed by allo-HSCT achieved a superior outcome compared with that of historical controls and results reported before the imatinib era [9], [10]. In almost all of these studies, matched related and matched unrelated donors were used for transplantation. We have had several reports from our center regarding haploidentical HSCT without in vitro T cell depletion that achieved promising long-term survival for patients, including both Ph+ ALL and Ph- ALL [11], [12]. However, no studies have documented the use of allo-HSCT with HID specifically for the treatment of children and adolescents with Ph+ ALL.

Therefore, in this study we aimed to explore the role of haploidentical HSCT for the treatment of pediatric patients with Ph+ ALL and investigated factors affecting the relapse rate and long-term survival of these children.

Section snippets

Patient eligibility

Pediatric patients between 1 and 18 years of age with Ph+ ALL who underwent haploidentical HSCT were consecutively enrolled. The diagnosis of Ph+ ALL was based on the World Health Organization diagnosis criteria [13]. All patients with ALL were screened for the diagnosis of Ph+ ALL using reverse transcription polymerase chain reaction (RT-PCR). The presence of the Philadelphia chromosome was also tested by standard karyotyping and/or FISH analysis for the BCR–ABL fusion gene. This study was

Enrollment

From December 2006 to March 2015, 58 consecutive pediatric patients with Ph+ ALL received allo-HSCT. Among these patients, 50 received HID transplant, and their median age was 13 years (range, 4–18 years). They received a median of six cycles of chemotherapy before HSCT (range, 3–14 cycles), and the median time from diagnosis to transplant was 180 days (range, 118–888 days). The demographic characteristics and relevant transplantation data of pediatric patients with Ph+ ALL undergoing HID

Discussion

In the imatinib era, the role of allo-HSCT for the treatment of pediatric patients with Ph+ ALL has been changing, largely because of results of the COG AALL003 study, which reported that allo-HSCT did not improve survival compared with intensive chemotherapy plus intensive imatinib therapy [7]. However, outcomes for pediatric patients who underwent HSCT in the EsPhALL study appeared superior to those in the non-transplant cohort, but limited by small number of patient without receiving

Conclusion

We conclude that HID HSCT could achieve promising long-term survival for pediatric patients with Ph+ ALL. BCR/ABL positivity post-transplant on +30 day was associated with increased risk for relapse after allo-HSCT in the imatinib era. There was some limitation in the current study including patients’ number. Carefully designed controlled study is needed to investigate whether HID transplant is preferable to children with BCR/ABL transcript positivity before transplant compared with intensive

Author contributions

H.C. designed the research, interpreted the data, and wrote the manuscript. K.-Y.L., P.-L.X., Y.-H.C., X.-H.Z., Y.W. performed the study and contributed to writing the manuscript. Y.-Z.Q. and Y.-R.L. performed the BCR-ABL RT-PCR and flow cytometry assays. Y.-Y. L. performed Karyotyping and FISH analysis. X.-J.H., the principal investigator, designed the research, interpreted the data, and wrote the manuscript. All authors read and approved the final manuscript.

Conflict of interest

There are no conflicts of interest to report.

Funding

This work was supported by grants from The Key Program of National Natural Science Foundation of China (No. 81230013) and from Scientific Research Foundation for Capital Medicine Development (No. CD 2016-1-4082).

Acknowledgement

We are grateful to Editage Group China for revisingand finalizing the manuscript.

References (27)

  • K.R.C.A. Schultz et al.

    Long-term follow-up of imatinib in pediatric Philadelphia chromosome-positive acute lymphoblastic leukemia: Children’s Oncology Group Study AALL0031

    Leukemia

    (2014)
  • M. Arico et al.

    Clinical outcome of children with newly diagnosed Philadelphia chromosome- positive acute lymphoblastic leukemia treated between 1995 and 2005

    J. Clin. Oncol.

    (2010)
  • S. Rives et al.

    Intermediate dose of imatinib in combination with chemotherapy followed by allogeneic stem cell transplantation improves early outcome in paediatric Philadelphia chromosome-positive acute lymphoblastic leukaemia (ALL): results of the Spanish Cooperative Group SHOP studies ALL-94, ALL-99 and ALL-2005

    Br. J. Haematol.

    (2011)
  • Cited by (9)

    • Safety and outcomes of maintenance therapy with third-generation tyrosine kinase inhibitor after allogeneic hematopoietic cell transplantation in Philadelphia chromosome positive acute lymphoblastic leukemia patients with T315I mutation

      2022, Leukemia Research
      Citation Excerpt :

      Supportive care was given as previously described [14]. During induction and consolidation therapy, first- or second-generation TKI was administered in combination with a standard dose of chemotherapy, as previously described [15,16]. If patients did not achieve adequate response to first-generation TKI, a second-generation TKI based on the results of the BCR-ABL mutation test was administered.

    • Prognosis of haploidentical hematopoietic stem cell transplantation in non-infant children with t(v;11q23)/MLL-rearranged B-cell acute lymphoblastic leukemia

      2020, Leukemia Research
      Citation Excerpt :

      Except for three patients (one receiving a matched sibling donor transplant [MSDT], and two with matched unrelated donor transplants [MUDT]), all remaining patients opted for haplo-HSCT. The preconditioning protocol was in accordance with previous reports [17,19–21]. The conditioning regimen for haplo-HSCT included cytarabine (4 g/m2/d)from days −10 to −9; busulfan (3.2 mg/kg/d) from days −8 to −6; Cy (1.8 g/m2/d) from days −5 to −4; simustine (250 mg/kg/d) on day −3, and anti-thymocyte globulin (ATG) (2.5 mg/kg/d; Sang Stat, Lyon, France) from days −5 to −2.

    • Allogeneic Hematopoietic Stem Cell Transplantation, Especially Haploidentical, May Improve Long-Term Survival for High-Risk Pediatric Patients with Philadelphia Chromosome–Positive Acute Lymphoblastic Leukemia in the Tyrosine Kinase Inhibitor Era

      2019, Biology of Blood and Marrow Transplantation
      Citation Excerpt :

      An unrelated cord blood (UCB) unit with a minimum of 4/6 HLA loci match transplantation was also considered as an alternative. Conditioning regimens were administered as previously described [12,14,16,18]. In MSD transplants the conditioning regimen was a modified busulfan-cyclophosphamide regimen, including the following: hydroxyurea (80 mg/kg/d) p.o. on day –10, Ara-c (2 g/m2/d) i.v. on day –9, busulfan (3.2 mg/kg/d) i.v. from days –8 to –6; cyclophosphamide (1.8 g/m2/d) i.v. from days –5 to –4; and methyl-N-(2-chloroethyl)-N-cyclohexyl-N-nitrosourea (250 mg/kg/d) p.o. on day –3.

    View all citing articles on Scopus
    View full text