Elsevier

The Lancet Oncology

Volume 19, Issue 12, December 2018, Pages 1617-1629
The Lancet Oncology

Articles
Interleukin 2 with anti-GD2 antibody ch14.18/CHO (dinutuximab beta) in patients with high-risk neuroblastoma (HR-NBL1/SIOPEN): a multicentre, randomised, phase 3 trial

https://doi.org/10.1016/S1470-2045(18)30578-3Get rights and content

Summary

Background

Immunotherapy with the chimeric anti-GD2 monoclonal antibody dinutuximab, combined with alternating granulocyte-macrophage colony-stimulating factor and intravenous interleukin-2 (IL-2), improves survival in patients with high-risk neuroblastoma. We aimed to assess event-free survival after treatment with ch14.18/CHO (dinutuximab beta) and subcutaneous IL-2, compared with dinutuximab beta alone in children and young people with high-risk neuroblastoma.

Methods

We did an international, open-label, phase 3, randomised, controlled trial in patients with high-risk neuroblastoma at 104 institutions in 12 countries. Eligible patients were aged 1–20 years and had MYCN-amplified neuroblastoma with stages 2, 3, or 4S, or stage 4 neuroblastoma of any MYCN status, according to the International Neuroblastoma Staging System. Patients were eligible if they had been enrolled at diagnosis in the HR-NBL1/SIOPEN trial, had completed the multidrug induction regimen (cisplatin, carboplatin, cyclophosphamide, vincristine, and etoposide, with or without topotecan, vincristine, and doxorubicin), had achieved a disease response that fulfilled prespecified criteria, had received high-dose therapy (busulfan and melphalan or carboplatin, etoposide, and melphalan) and had received radiotherapy to the primary tumour site. In this component of the trial, patients were randomly assigned (1:1) to receive dinutuximab beta (20 mg/m2 per day as an 8 h infusion for 5 consecutive days) or dinutuximab beta plus subcutaneous IL-2 (6 × 106 IU/m2 per day on days 1–5 and days 8–12 of each cycle) with the minimisation method to balance randomisation for national groups and type of high-dose therapy. All participants received oral isotretinoin (160 mg/m2 per day for 2 weeks) before the first immunotherapy cycle and after each immunotherapy cycle, for six cycles. The primary endpoint was 3-year event-free survival, analysed by intention to treat. This trial was registered with ClinicalTrials.gov, number NCT01704716, and EudraCT, number 2006-001489-17, and recruitment to this randomisation is closed.

Findings

Between Oct 22, 2009, and Aug 12, 2013, 422 patients were eligible to participate in the immunotherapy randomisation, of whom 406 (96%) were randomly assigned to a treatment group (n=200 to dinutuximab beta and n=206 to dinutuximab beta with subcutaneous IL-2). Median follow-up was 4·7 years (IQR 3·9–5·3). Because of toxicity, 117 (62%) of 188 patients assigned to dinutuximab beta and subcutaneous IL-2 received their allocated treatment, by contrast with 160 (87%) of 183 patients who received dinutuximab beta alone (p<0·0001). 3-year event-free survival was 56% (95% CI 49–63) with dinutuximab beta (83 patients had an event) and 60% (53–66) with dinutuximab beta and subcutaneous IL-2 (80 patients had an event; p=0·76). Four patients died of toxicity (n=2 in each group); one patient in each group while receiving immunotherapy (n=1 congestive heart failure and pulmonary hypertension due to capillary leak syndrome; n=1 infection-related acute respiratory distress syndrome), and one patient in each group after five cycles of immunotherapy (n=1 fungal infection and multi-organ failure; n=1 pulmonary fibrosis). The most common grade 3–4 adverse events were hypersensitivity reactions (19 [10%] of 185 patients in the dinutuximab beta group vs 39 [20%] of 191 patients in the dinutuximab plus subcutaneous IL-2 group), capillary leak (five [4%] of 119 vs 19 [15%] of 125), fever (25 [14%] of 185 vs 76 [40%] of 190), infection (47 [25%] of 185 vs 64 [33%] of 191), immunotherapy-related pain (19 [16%] of 122 vs 32 [26%] of 124), and impaired general condition (30 [16%] of 185 vs 78 [41%] of 192).

Interpretation

There is no evidence that addition of subcutaneous IL-2 to immunotherapy with dinutuximab beta, given as an 8 h infusion, improved outcomes in patients with high-risk neuroblastoma who had responded to standard induction and consolidation treatment. Subcutaneous IL-2 with dinutuximab beta was associated with greater toxicity than dinutuximab beta alone. Dinutuximab beta and isotretinoin without subcutaneous IL-2 should thus be considered the standard of care until results of ongoing randomised trials using a modified schedule of dinutuximab beta and subcutaneous IL-2 are available.

Funding

European Commission 5th Frame Work Grant, St. Anna Kinderkrebsforschung, Fondation ARC pour la recherche sur le Cancer.

Introduction

Neuroblastoma is a paediatric extracranial malignancy of the sympathetic nervous system that is responsible for 12% of childhood cancer deaths.1, 2 High-risk neuroblastoma, which is defined by the presence of metastatic disease in children older than 12 or 18 months2 or the MYCN amplification (MNA) in patients of any age, is associated with 5-year survival of only 40%.3, 4 Current therapeutic approaches consist of intensive induction,5, 6 high-dose chemotherapy, and autologous stem-cell rescue as consolidation treatment,4, 7 and isotretinoin for minimal residual disease therapy. Treatment with the human and mouse chimeric antibody ch14.18, directed against disialoganglioside GD2, has been developed as an important option in the treatment phase for patients with minimal residual disease.8, 9 GD2 is expressed on most neuroblastoma cells, and in normal human tissues is only expressed by neurons, skin melanocytes, and peripheral sensory nerve fibres, which make GD2 a suitable target for immunotherapy.8

In Europe, ch14.18 was recloned in Chinese hamster ovary (CHO) cells (dinutuximab beta)10 and made available for clinical trials of the International Society of Paediatric Oncology Europe Neuroblastoma (SIOPEN) group. A phase 1 study of dinutuximab beta11 showed tolerability and activity in a regimen of 20 mg/m2 given by an 8 h infusion on 5 consecutive days.

Research in context

Evidence before this study

Preclinical and preliminary clinical data indicate that monoclonal antibodies against the tumour-associated disialoganglioside GD2 have activity against neuroblastoma, which was enhanced when the monoclonal antibody (ch14.18) was combined with either granulocyte-macrophage colony- stimulating factor (GM-CSF) or interleukin 2 (IL-2). A randomised front-line trial in patients with high-risk neuroblastoma comparing the addition of ch14.18 derived from SP2/0 hybridoma cells (ch14.18/SP2/0, dinutuximab), GM-CSF, and IL-2 to standard of care (oral isotretinoin), versus isotretinoin alone in residual disease therapy, showed a survival benefit in the immunotherapy group. However, the role of cytokines in this context remained unclear. The International Society of Paediatric Oncology European Neuroblastoma Group recloned ch14.18 in Chinese hamster ovary cells (ch14.18/CHO, dinutuximab beta) and investigated its safety and tolerability. Because IL-2 has the ability to increase the number of natural killer cells and to enhance antibody-dependent cell-mediated cytotoxicity, we decided to study the combination of IL-2 with dinutuximab beta to assess the effect on survival. A feasible, safe, and effective dose for subcutaneous IL-2 had been established in a phase 1/2 trial. We searched PubMed on Jan 15, 2002, and Oct 1, 2007, for all publications with the terms “neuroblastoma”, “randomised”, and “chimeric anti-GD2 antibody” with no language or date restrictions. We found one trial, which compared the addition of a monoclonal antibody against GD2, added to chemotherapy in the relapse setting. We did not find any randomised trials comparing different immunotherapy regimens in front-line patients during residual disease.

Added value of this study

The addition of subcutaneous IL-2 (6 x 106 IU/m2) to dinutuximab beta (5 x 20 mg/m2 per day, in an 8 h infusion) in patients with high-risk neuroblastoma who had responded to induction and consolidation treatment did not increase event-free survival or overall survival, suggesting that this combination in this schedule has no therapeutic benefit. Furthermore, we noted an increased inflammatory toxicity profile in the group receiving IL-2. Dinutuximab beta with isotretinoin was associated with objective responses in patients with residual, evaluable disease. This study adds information to optimise therapeutic schedules that use monoclonal antibodies against GD2, and highlights the respective contributions of cytokine and antibody to efficacy and toxicity.

Implications of all the available evidence

Monoclonal antibodies against the tumour-associated disialoganglioside GD2 improve survival in patients with high-risk neuroblastoma. However, the addition of subcutaneous IL-2 to dinutuximab beta did not improve outcomes and increased toxicity. Further clinical trials of different dosing schedules and combinations are required to optimise the benefit of immunotherapy with anti-GD2 monoclonal antibodies in high-risk neuroblastoma.

On March 15, 2006, SIOPEN started a randomised trial (HR-NBL1/SIOPEN) comparing dinutuximab beta and isotretinoin with isotretinoin alone in children with high-risk neuroblastoma who had received dose-intensive induction, surgical excision of the primary tumour, high-dose chemotherapy followed by autologous stem-cell rescue, and local radiotherapy. However, on April 15, 2007, the Children's Oncology Group first became aware of the early results of their ANBL0032 study (NCT00026312) and later reported that the addition of a ch14.18 antibody produced in SP2/0 cells (dinutuximab), combined with cytokines (granulocyte–macrophage colony-stimulating factor [GM-CSF] and interleukin 2 [IL-2]), in addition to isotretinoin, improved 2-year event-free survival by 20% and overall survival by 11% in patients with high-risk neuroblastoma.8 In this three-drug regimen the individual contribution to efficacy of each of the drugs was unknown, and the combination was associated with substantial toxicity, including pain, fever, allergic reactions, and capillary leak syndrome.

In view of these results, continuation of the SIOPEN trial comparing dinutuximab beta and isotretinoin to isotretinoin alone was deemed neither feasible nor ethical. Clinical and preclinical data suggested that IL-2 in addition to ch14.18 had a positive immunomodulatory effect.12 Because subcutaneous IL-2 (6 × 106 IU/m2 per day) was associated with mild toxicity and little discomfort in adults and could be administered in an outpatient setting,13 SIOPEN adopted this schedule in a dose-finding study and showed that the regimen of 6 × 106 IU/m2 per day of subcutaneous IL-2 was acceptable in children with high-risk neuroblastoma when administered on 5 consecutive days in five intermittent courses over 3 months.14 Additionally, GM-CSF was neither licensed nor available in Europe, so the HR-NBL1/SIOPEN trial was amended to randomly assign patients to receive either dinutuximab beta and subcutaneous IL-214 with oral isotretinoin or dinutuximab beta with oral isotretinoin, to investigate the role of subcutaneous IL-2 in improving event-free survival in children with high-risk neuroblastoma.

Section snippets

Study design and participants

The HR-NBL1/SIOPEN trial is an international, randomised, open-label, phase 3 trial. The protocol is available online.

Recruitment to all randomised study groups is closed. Results from two randomisations have been published (prophylactic granulocyte colony-stimulating factor during induction; busulfan and melphalan vs carboplatin, etoposide, and melphalan as high-dose chemotherapy6, 7), with two more awaiting data maturity before publication (continuous infusion of dinutuximab beta with or

Results

Between Oct 22, 2009, and Aug 12, 2013, 422 (68%) of 621 patients who had been recruited at SIOPEN member centres and were participating in the HR-NBL1/SIOPEN trial were eligible for random allocation to immunotherapy, of whom 406 (96%) were randomly assigned (n=200 to receive dinutuximab beta, n=206 to receive dinutuximab beta with subcutaneous IL-2; figure 1; appendix pp 2–5). Assignment to immunotherapy was closed after the calculated sample size was reached and data were released by the

Discussion

In this open-label, phase 3, randomised trial, we compared dinutuximab beta and oral isotretinoin to dinutuximab beta combined with subcutaneous IL-2 and oral isotretinoin in patients with high-risk neuroblastoma who responded to induction and consolidation therapy. The addition of subcutaneous IL-2 to dinutuximab beta, in the dose and schedule administered, did not increase event-free survival or overall survival and was associated with increased toxicity.

Dinutuximab (ch14.18/SP2/0) in

References (30)

  • N-K V Cheung et al.

    Neuroblastoma: developmental biology, cancer genomics and immunotherapy

    Nat Rev Cancer

    (2013)
  • R Ladenstein et al.

    Ch14.18 antibody produced in CHO cells in relapsed or refractory stage 4 neuroblastoma patients: a SIOPEN phase 1 study

    MAbs

    (2013)
  • BM Mueller et al.

    Enhancement of antibody-dependent cytotoxicity with a chimeric anti-GD2 antibody

    J Immunol

    (1990)
  • NJ Meropol et al.

    Daily subcutaneous injection of low-dose interleukin 2 expands natural killer cells in vivo without significant toxicity

    Clin Cancer Res

    (1996)
  • R Ladenstein et al.

    Dose finding study for the use of subcutaneous recombinant interleukin-2 to augment natural killer cell numbers in an outpatient setting for stage 4 neuroblastoma after megatherapy and autologous stem-cell reinfusion

    J Clin Oncol

    (2011)
  • Cited by (252)

    • Precision Oncology in Pediatric Cancer Surgery

      2024, Surgical Oncology Clinics of North America
    View all citing articles on Scopus
    *

    Joint first authors

    For the International Society of Paediatric Oncology European Neuroblastoma Group (SIOPEN)

    3

    Dr Brock retired in April, 2014.

    4

    Dr Holmes retired in September, 2011.

    5

    Dr Loibner retired in June, 2018.

    6

    Prof Pearson retired in May, 2015.

    7

    Prof Yaniv retired in February, 2018.

    View full text