A Treatment Study Protocol for Participants 0-45 Years With Acute Lymphoblastic Leukaemia

Sponsor
Mats Heyman (Other)
Overall Status
Recruiting
CT.gov ID
NCT04307576
Collaborator
The Swedish Research Council (Other), The Swedish Childhood Cancer Foundation (Other), Pfizer (Industry), Servier (Industry), NordForsk (Other), Aamu Pediatric Cancer Foundation (Other), Gesellschaft fur Padiatrische Onkologie und Hamatologie - Germany (Other), Clinical Trial Center North (CTC North GmbH & Co. KG) (Other), Belgium Health Care Knowledge Centre (Other), Karolinska Institutet (Other), Cancer Research UK (Other), Fundação Rui Osório de Castro (Other), Acreditar - Associação de Pais e Amigos das Crianças com Cancro (Other), Grupo Português De Leucemias Pediátricas (Other), Amgen (Industry), Nova Laboratories Limited (Industry), Danish Child Cancer Foundation (Other), Danish Cancer Society (Other), The Novo Nordic Foundation (Other)
6,430
101
10
143.6
63.7
0.4

Study Details

Study Description

Brief Summary

ALLTogether collects the experience of previously successful treatment of infants, children and young adults, with ALL from a number of well-renowned study groups into a new master protocol, which is both a comprehensive system for stratification and treatment of ALL in this age-group as well as the basis for several randomised and interventional trials included in the study-design.

Condition or Disease Intervention/Treatment Phase
  • Drug: Omitted Doxorubicin
  • Drug: Omitted Vincristine+Dexamethasone pulses
  • Drug: Inotuzumab Ozogamicin+Standard Maintenance Therapy
  • Drug: Imatinib
  • Drug: 6-tioguanine+Standard Maintenance Therapy
  • Drug: Blinatumomab
Phase 3

Detailed Description

ALLTogether is a European clinical treatment study for acute lymphoblastic leukaemia (ALL) in infants, children and young adults. The aims are to improve survival and quality of survival. In young people, ALL has excellent outcome with an overall survival of about 92% in children and 75% in young adults. Infants with BCP-ALL and KMT2A-rearrangements have a worse outcome and are treated according to separate protocols, but infants with KMT2A-germline and T-cell ALL have acceptable outcome on standard ALL therapy. However, patients still die of disease - from relapse because of under-treatment and a large fraction of patients are also over-treated: All patients risk treatment-related death and some suffer long-term side-effects or secondary cancer. To show improvement with such good survival, large populations are needed.

Study groups from Sweden, Norway, Iceland, Denmark, Finland, Estonia and Lithuania (NOPHO), the UK (UKALL), the Netherlands (DCOG), Germany (COALL), Belgium (BSPHO), Portugal (SHOP), Ireland (PHOAI), and France (SFCE), have designed a common treatment protocol.

The study has a complex clinical trial design with sub-protocols (the randomisations / intervention) connected to a master protocol. The master protocol consists of well established therapy-elements and in its design typical for current ALL therapy. The master protocol therapy is in the study design considered as standard of care (SOC) therapy for infants, children and young adults with ALL.

The study structure is defined by a master protocol onto which randomised and interventional sub-protocols as well as sub-studies may be added, run and stop in a modular fashion.

The randomisations / intervention may identify therapy that is less toxic, but equally efficacious for sub-groups of patients and innovative therapy that may reduce relapses and death from ALL. In the master protocol, improved risk-stratification is likely to increase survival and reduce unnecessary toxicity and the introduction of therapeutic drug monitoring (TDM) of Asparaginase activity will make the use of Asparaginase more rational and efficient and may thus improve overall outcomes.

The investigators hypothesise that patients stratified to the standard-risk group are over-treated. Therefore, it will be tested if the treatment can be safely reduced. In the R1 randomisation, patients will be randomised to receiving the Delayed Intensification (DI) phase of therapy with or without the anthracycline Doxorubicin.

A similar hypothesis of over-treatment will also be tested in patients stratified to the intermediate risk-low group. In the R2 randomisation patients will be randomly assigned to either removal of Doxorubicin during the DI phase or removal of Vincristine and Dexamethasone pulses during the maintenance phase or to the control group, which will be treated with Doxorubicin in DI as well as Vincristine and Dexamethasone pulses during maintenance. Patients will only be randomised once.

Randomisation R1 and R2 are only considered for children since adults have worse outcome and very poor survival after relapse, but the risk-stratification is likely to reduce the number of high-risk cases also in the adult-group.

Patients stratified as intermediate risk-high (IR-high) are identified as having an increased risk of relapse and thus a less favourable prognosis than the standard- and intermediate risk-low groups, but a more favourable prognosis than the high risk patients. The majority of all relapses in childhood ALL is expected to occur in the IR-high group. Following a relapse, only approximately 40% of the children can be successfully treated again and for adults the corresponding figure is less than 20 %, so preventing relapses is very important. New treatment options that improves the antileukaemic efficacy and which have an improved safety profile are urgently needed.

For IR-high patients Randomisation 3 (R3) is available. In R3 patients will be randomised to receive either:

  1. the addition of two cycles of Inotuzumab ozogamicin (InO) - Besponsa®, before start of the maintenance phase. After these cycles, the patients randomised to the InO arm will receive maintenance for the same duration as in the control arm.

  2. the addition of low dose 6-tioguanine (6TG) as an addition to the standard maintenance therapy.

  3. standard maintenance therapy

Patients with ABL-class fusions in their leukaemic clone will, as a non-randomised experimental intervention, be treated with an addition of a tyrosine-kinase inhibitor during the induction phase (for patients <25 years) and from the consolidation phase (patients ≥25 years). This intervention may shift therapy for previously resistant cases to lower intensity treatment with the associated reduced morbidity and may also reduce the number of relapses in analogy with the results in Ph+ ALL. The reason for not performing a randomised comparison is the rarity of the aberration and also the diversity of ABL-class fusions, reducing statistical power for any comparison further. For this reason, the results of this intervention may be pooled with other study-groups trying similar approaches.

A new intervention is introduced for Down syndrome patients with CD19 positive ALL:

ALLTogether1 DS (NRI2). For Down syndrome-ALL patients who have end of Induction MRD detectable but <25% two conventional chemotherapy consolidation blocks will be replaced with two blocks of Blinatumomab.

For high-risk B-lineage patients, CAR-T therapy can be an alternative to high-risk blocks and stem-cell transplant, but in this case the intervention (CAR-T infusion) will be performed outside the ALLTogether1 study. However, the stratification-system in ALLTogether1 will define the population with a potential CAR-T indication.

ALLTogether1 also includes five sub-studies:

Efficacy and pharmacokinetics of Imatinib in ABL-class fusion positive ALL

Target population: All ABL-class patients enrolled in the ALLTogether study. Biomaterials to be collected at diagnosis, during therapy, follow-up and relapse.

Aims

  1. To determine the efficacy of imatinib in the treatment of ABL-class leukemia

  2. To find the best discriminative biomarkers for TKI response in ABL-class ALL

  3. To determine the frequency of intrinsic (at diagnosis) and acquired TKI resistance (due to treatment)

  4. To find causes of TKI resistance in ABL-class patients

  5. To describe the pharmacokinetics of Imatinib in TKI-treated patients

Objectives

  1. To determine the percent of ABL-class patients who need to switch from IR-high to HR because of high MRD levels

  2. To determine the effect of imatinib exposure on clinical outcome, including pharmacokinetic measurements of imatinib

  3. To determine the molecular response to imatinib by monitoring fusion transcript levels and mutational spectrum at diagnosis and during follow up

  4. To determine whether the molecular response parameters reflect the Ig/TCR MRD or flow-MRD response or are a better predictor of therapy failure than Ig/TCR or flow-based MRD monitoring

  5. To determine the phosphorylation status of ABL-class proteins and presence of TKI-resistance associated mutations in ABL genes prior to imatinib treatment and the emergence of such mutations during treatment with imatinib

  6. To determine the presence of mutations in regulatory /other genes before and during imatinib treatment and functionally address the importance of these mutations in TKI resistance

  7. To determine whether the efficacy of TKIs depends on the type of fusion gene

Biomarkers to Reform Approaches to therapy-Induced Neurotoxicity (BRAIN)

Target population: All patients registered on ALLTogether1 aged ≥ 4 years at end of therapy and without:

  1. Pre-existing neurodevelopmental disorder (e.g Trisomy 21, ADHD) prior to diagnosis of ALL

  2. Significant visual or motor impairment preventing use of a touch screen ipad

Aims

  1. To institute universal screening of all children for adverse neurocognitive outcomes at the end of treatment using a validated user-friendly computer software programme (CogState) and compare neurocognitive outcomes by treatment allocation.

  2. To identify risk factors for adverse outcomes including whether acute neurotoxic events are associated with poor performance on cognitive tests at end of therapy compared to patients without acute neurotoxicity.

Primary end-point

  1. Proportion of children with a z-score <1.5 on detection and/or identification CogState tasks in each treatment arm at the end of anti-leukaemic therapy. A z-score < 1.5 correlates with moderate cognitive impairment at a level that may require additional support.

Secondary and exploratory end-points

  1. Association between CogState scores at end of treatment and overt neurotoxic episodes as recorded on the trial adverse event database.

  2. Association between Cogstate scores and clinical and demographic variables - age, sex, ethnicity, CNS status.

  3. Proportion of children with scores <1.5SD for one card learning (learning), one back (working memory) and Groton's maze (executive function) on different treatment arms.

  4. Association between CogState scores and patient reported outcome measures/Quality of life measurements collected as part of the main ALLTogether1 trial.

Association between asparaginase activity levels and outcome

Target population: All patients included in the ALLTogether1 protocol are eligible for participation.

Primary aim

To study the association between asparaginase activity levels and outcome (MRD, relapse, survival)

Secondary aims

  1. To evaluate the association between asparaginase activity levels and toxicities, such as pancreatitis, infections and deep venous thrombosis (DVT)

  2. To evaluate the association between asparaginase activity levels and hepatotoxicity in a subset of patients

CSF-Flow

Target population: All patients included in the ALLTogether1 protocol are eligible for participation

Aims

  1. To use cerebrospinal fluid (CSF) flow cytometry (FCM) to improve the accuracy of diagnostic tests for CNS leukaemia compared to conventional CSF cytology. An associated objective will be to develop a recommended protocol for CSF flow cytometry with external quality assessment to ensure uniformity of measurement across the ALLTogether consortium.

  2. To investigate whether negative FCM identifies a group of children at very low risk of CNS relapse, suitable for testing de-escalation of CNS-directed therapy in future trials.

  3. To investigate whether positive FCM can identify children at increased risk of CNS relapse and whether patients with persistent positivity (FCM positive at day 15 onwards) might benefit from studies testing escalated CNS-directed therapy or a switch to more intensive treatment arms.

  4. To collect matching CSF supernatant for studies comparing CSF FCM with soluble biomarkers (e.g. metabolic, cell-free DNA, proteomic and microRNA) in selected centres.

Maintenance therapy pharmacokinetics/-dynamics study

Target population: All patients included in the ALLTogether1 protocol are eligible for participation. For IR-high patients participating in the randomised InO- and TEAM sub-protocols, the monitoring of 6-mercaptopurine (6MP)/Methotrexate (MTX) metabolites at three months intervals is mandatory.

Aims and specific objectives

  1. To map pharmacokinetics of 6MP and MTX during maintenance therapy in all patients in the ALLTogether protocol.

  2. To associate metabolite profiles with TPMT and NUDT15 variants, as routinely analysed in ALLTogether.

  3. To explore the association of event-free survival with DNA-TG and other 6MP/MTX metabolites.

  4. To explore the association between risk of second cancers with DNA-TG and other 6MP/MTX metabolites.

  5. To explore the association of risk of invasive infections with DNA-TG and other 6MP/MTX metabolites.

  6. To explore the association of risk of osteonecrosis with DNA-TG and other 6MP/MTX metabolites.

  7. To explore the association of sinusoidal obstruction syndrome with DNA-TG and other 6MP/MTX metabolites.

Study Design

Study Type:
Interventional
Anticipated Enrollment :
6430 participants
Allocation:
Randomized
Intervention Model:
Sequential Assignment
Intervention Model Description:
Master protocol with risk-stratification. The risk-stratified groups will proceed to non-randomised or randomised interventions in single arm (TKI) and (Blinatumomab) and parallel (R1/R2/R3) models respectively.Master protocol with risk-stratification. The risk-stratified groups will proceed to non-randomised or randomised interventions in single arm (TKI) and (Blinatumomab) and parallel (R1/R2/R3) models respectively.
Masking:
None (Open Label)
Primary Purpose:
Treatment
Official Title:
ALLTogether1 - A Treatment Study Protocol of the ALLTogether Consortium for Children and Young Adults (0-45 Years of Age) With Newly Diagnosed Acute Lymphoblastic Leukaemia (ALL)
Actual Study Start Date :
Jul 13, 2020
Anticipated Primary Completion Date :
Jun 30, 2027
Anticipated Study Completion Date :
Jun 30, 2032

Arms and Interventions

Arm Intervention/Treatment
No Intervention: R1 - SR standard arm

Standard risk arm receiving standard treatment (Delayed Intensification including Doxorubicin).

Experimental: R1 - SR experimental arm

Standard risk arm, receiving Delayed Intensification without Doxorubicin IV 3 x 30 mg/m2/dose.

Drug: Omitted Doxorubicin
Omission of IV Doxorubicin

No Intervention: R2 - IR-low standard arm

Standard treatment with Delayed Intensification including Doxorubicin and Maintenance including Vincristine+Dexamethasone pulses.

Experimental: R2 - IR-low experimental arm A

Standard treatment with omission of Doxorubicin IV 3 x 30 mg/m2/dose in the Delayed Intensification phase.

Drug: Omitted Doxorubicin
Omission of IV Doxorubicin

No Intervention: R3 - IR-high standard arm

Intermediate risk high arm receiving Standard Maintenance Therapy.

Experimental: R3-InO - IR-high experimental arm

Inotuzumab IV 0,5 mg/m2, given on days 253, 260, 267 and on days 274, 281, 288 before start of Standard Maintenance Therapy.

Drug: Inotuzumab Ozogamicin+Standard Maintenance Therapy
Addition of IV Inotuzumab ozogamicin before Maintenance Therapy
Other Names:
  • Besponsa+Maintenance Therapy
  • Experimental: ABL-class fusions intervention

    Imatinib p.o. 340 mg/m2 given daily from day 15 or 30 (depending on age) to the end of therapy (week 106) in addition to Standard IR-high chemotherapy.

    Drug: Imatinib
    p.o. Imatinib

    Experimental: R3-TEAM - IR-high experimental arm

    6-tioguanine p.o, 2,5-12,5 mg/m2, given daily in addition to Standard Maintenance Therapy.

    Drug: 6-tioguanine+Standard Maintenance Therapy
    Addition of p.o. 6-tioguanine to Standard Maintenance Therapy

    Experimental: ALLTogether1 DS Blinatumomab intervention

    Blinatumomab IV, 5 mcg/m2/day up to 28 mcg/day (detailed dosing in protocol) continous infusion. Two 28 day courses with a two week treatment free interval in between. Blinatumomab courses replace Consolidation 1 and Consolidation 2 in the standard protocol adapted for Down syndrome patients.

    Drug: Blinatumomab
    IV Blinatumomab
    Other Names:
  • Blincyto
  • Experimental: R2 - IR-low experimental arm B

    Standard treatment with omission of monthly pulses of Vincristine IV 1,5 mg/m2/dose and 5 days of Dexamethasone p.o. 6 mg/m2/day in the Maintenance Phase.

    Drug: Omitted Vincristine+Dexamethasone pulses
    Omission of Vincristine+Dexamethasone pulses

    Outcome Measures

    Primary Outcome Measures

    1. Event-free survival (EFS) for the whole protocol [5 year estimates from the time of diagnosis will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up.]

      The primary endpoint for the whole protocol (compared with the legacy protocols of the participating study-groups forming the consortium) is event-free survival (EFS) - as defined in the protocol.

    2. Event-free survival (EFS) for the TKI intervention [From the start of TKI (day 15 or day 30), 5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up for all interventions except Inotuzumab-randomisation (minimum 2-year follow-up).]

      The primary endpoint for the TKI intervention is event-free survival (EFS) - as defined in the protocol, from the start of TKI until event or end of follow-up

    3. Disease-free survival (DFS) R1 + R2 [5 and 8 year estimates from the time of randomisation will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up.]

      The primary endpoint for Randomisation 1 and 2 is disease-free survival (DFS) - as defined in the protocol counting from the time of randomisation

    4. Disease-free survival (DFS) R3 [5 year estimates from the time of randomisation will be measured but adequate follow-up for these estimates will be ensured: at least 2-year follow-up]

      The primary endpoint for Randomisation 3 and the ABL-class fusion intervention is disease-free survival (DFS) - as defined in the protocol counting from the time of randomisation (R3) and the start of TKI-therapy (ABL-class fusion intervention).

    5. MRD response after 1 cycle of Blinatumomab [End of first Blinatumomab infusion +/- 1 week]

      Fraction of patients with undetectable MRD ("Complete MRD response") at the end of one cycle of Blinatumomab (+/- 1 week)

    Secondary Outcome Measures

    1. Overall survival (OS) for the whole protocol [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up.]

      Overall survival defined as time from diagnosis to death or end of follow-up for surviving patients.

    2. Overall survival (OS) for R1 + R2 [5 and 8 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up.]

      Overall survival defined as time from randomisation to death or end of follow-up for surviving patients.

    3. Overall survival (OS) for R3 [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 2 years follow-up]

      Overall survival defined as time from randomisation to death or end of follow-up for surviving patients.

    4. Overall survival (OS) for R3-TEAM associated with DNA-TG [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 2 years follow-up]

      Overall survival as defined above in relation to DNA-TG.

    5. Overall survival (OS) for TKI [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up (TKI).]

      Overall survival defined as time from start of TKI to death or end of follow-up for surviving patients

    6. Overall survival (OS) for ALLTogether1 DS [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up.]

      Overall survival defined as time from start of Blinatumomab to death or end of follow-up for surviving patients

    7. Induction death [From diagnosis until death before remission (at the earliest, day 29) or in case of no CR day 29, completion of induction and consolidation 1 (protocol day 99 - Down) and in addition 3 high-risk blocks (protocol day 134 - all other patients)]

      Fraction of patients who die as well as cumulative incidence of death before achieved complete remission (CR) within the time-frame described in the protocol

    8. Resistant disease [From diagnosis until achieved complete remission (at the earliest, day 29) or in case of no CR day 29, assessment after induction and consolidation 1 (protocol day 99-Down patients) and in addition 3 high-risk blocks (protocol day 134-all other patients)]

      Fraction of patients as well as cumulative incidence of resistant disease as described in the protocol. Induction death as competing risk.

    9. Cumulative incidence of relapse for the whole protocol [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up.]

      Time from achieved complete remission until relapse as defined in the protocol or end of follow-up. Second malignancy, death in complete remission as competing events.

    10. Cumulative incidence of relapse for R1 + R2 [5 and 8 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up.]

      Time from randomisation until relapse as defined in the protocol or end of follow-up. Second malignancy, death in complete remission as competing events.

    11. Cumulative incidence relapse for R3 [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 2 years follow-up]

      Time from randomisation until relapse as defined in the protocol or end of follow-up. Second malignancy, death in complete remission as competing events.

    12. Cumulative incidence relapse for R3-TEAM in association with DNA-TG [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 2 years follow-up]

      Cumulative incidence of relapse as defined for R3 in association with DNA-TG for R3-TEAM. Second malignancy, death in complete remission as competing events.

    13. Cumulative incidence CD22 negative relapse for R3 [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 2 years follow-up]

      Time from randomisation until relapse without expression of CD22 as defined in the protocol or end of follow-up. Second malignancy, death in complete remission and relapse with CD22 expression as competing events.

    14. Cumulative incidence relapse for TKI [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up]

      Time from start of TKI until relapse as defined in the protocol or end of follow-up. Second malignancy, death in complete remission as competing events.

    15. Cumulative incidence relapse for ALLTogether1 DS [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up]

      Time from start of Blinatumomab until relapse as defined in the protocol or end of follow-up. Second malignancy, death in complete remission as competing events.

    16. Cumulative incidence of CD19 negative relapse for ALLTogether1 DS [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up]

      Time from start of Blinatumomab until CD19 negative relapse as defined in the protocol or end of follow-up. Second malignancy, death in complete remission and CD19 positive relapse as competing events.

    17. Cumulative incidence of second malignant neoplasm (SMN) for the whole protocol [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up.]

      Time from achieved complete remission until diagnosis of second malignant neoplasm as defined in the protocol or end of follow-up. Relapse and death in complete remission as competing events.

    18. Cumulative incidence of second malignancy for R1+R2 [5 and 8 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up.]

      Time from randomisation until diagnosis of second malignant neoplasm as defined in the protocol or end of follow-up. Relapse and death in complete remission as competing events.

    19. Cumulative incidence of second malignancy for R3 [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 2 years follow-up.]

      Time from randomisation until diagnosis of second malignant neoplasm as defined in the protocol or end of follow-up. Relapse and death in complete remission as competing events.

    20. Cumulative incidence of second malignancy for R3-TEAM in association with DNA-TG [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 2 years follow-up.]

      Cumulative incidence of second malignancy as defined above for R3 in association with DNA-TG for R3-TEAM. Relapse and death in complete remission as competing events.

    21. Cumulative incidence of second malignancy for TKI [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up.]

      Time from start of TKI until diagnosis of second malignant neoplasm as defined in the protocol or end of follow-up. Relapse and death in complete remission as competing events.

    22. Cumulative incidence of second malignancy for ALLTogether1 DS [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up.]

      Time from start of Blinatumomab until diagnosis of second malignant neoplasm as defined in the protocol or end of follow-up. Relapse and death in complete remission as competing events.

    23. Cumulative incidence of death in complete remission for the whole protocol [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up.]

      Time from achieved complete remission until death in complete remission as defined in the protocol or end of follow-up. Relapse and second malignant neoplasm as competing events.

    24. Cumulative incidence of death in complete remission for R1+R2 [5 and 8 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 2 years follow-up.]

      Time from randomisation until death in complete remission as defined in the protocol or end of follow-up. Relapse and second malignant neoplasm as competing events.

    25. Cumulative incidence of death in complete remission for R3 [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 2 years follow-up]

      Time from randomisation until death in complete remission as defined in the protocol or end of follow-up. Relapse and second malignant neoplasm as competing events.

    26. Cumulative incidence of death in complete remission for TKI [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up]

      Time from start of TKI until death in complete remission as defined in the protocol or end of follow-up. Relapse and second malignant neoplasm as competing events.

    27. Cumulative incidence of death in complete remission for ALLTogether1 DS [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up]

      Time from start of Blinatumomab until death in complete remission as defined in the protocol or end of follow-up. Relapse and second malignant neoplasm as competing events.

    28. Cumulative incidence of treatment-related mortality for the whole protocol [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up.]

      Time from diagnosis until death during induction, death in complete remission or death from second malignant neoplasm (if not related to SMN-treatment).

    29. Cumulative incidence of treatment-related mortality R1+R2 [5 and 8 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 2 years follow-up.]

      Time from randomisation until death in complete remission or death from second malignant neoplasm (if not related to SMN-treatment).

    30. Cumulative incidence of treatment-related mortality R3 [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 2 years follow-up]

      Time from randomisation until death in complete remission or death from second malignant neoplasm (if not related to SMN-treatment).

    31. Cumulative incidence of treatment-related mortality TKI [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up]

      Time from start of TKI until death in complete remission or death from second malignant neoplasm (if not related to SMN-treatment).

    32. Leukaemia specific mortality for the whole protocol [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up.]

      Time from diagnosis until death after resistant disease or relapse - as defined in the protocol.

    33. Leukaemia specific mortality for R1+R2 [5 and 8 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up.]

      Time from randomisation until death after relapse - as defined in the protocol.

    34. Leukaemia specific mortality for R3 [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 2 years follow-up]

      Time from randomisation until death after relapse - as defined in the protocol.

    35. Leukaemia specific mortality for TKI [5 year estimates will be measured but adequate follow-up for these estimates will be ensured: at least 5 years follow-up]

      Time from start of TKI until death after relapse - as defined in the protocol.

    36. Incidence of Adverse Events of Special Interest (AESIs) per treatment-phase in the whole protocol and TKI intervention [From time of diagnosis after each treatment-phase (one extra in the middle of maintenance) and annually until 5 years from discontinuation of therapy.]

      Cumulative incidence of 19 AESIs as defined in the protocol

    37. Incidence of Adverse Events of Special Interest (AESIs) extra assessment (R1+R2) [Cumulative incidence of AESIs estimated 3 months after start of maintenance (R1+R2) and at the end of maintenance (R2)]

      Cumulative incidence of 4 additional AESIs as defined in the protocol

    38. Incidence of Adverse Events of Special Interest (AESIs) extra assessment (R3) [Cumulative incidence of AESIs estimated at the end of maintenance.]

      Cumulative incidence of 3 additional AESIs as defined in the protocol

    39. Incidence of Serious Adverse Events (SAEs) and Adverse Events (AEs) related to R3 [From time of randomisation until the end of maintenance therapy (approximately 77 weeks from randomisation)]

      Cumulative incidence of SAEs and AEs (with limitations) as defined in the protocol

    40. Quantitative measures of toxicity R1+R2 [From time of randomisation, assessment after delayed intensification and 6 weeks after start of maintenance]

      Days: in hospital, with iv antibiotics, with iv analgesics, with iv nutritional support

    41. Metabolic consequences of steroid exposure (R2) [At the end of therapy (approximately 94 weeks from randomisation) and 5 years after discontinuation of treatment]

      Measurements of BMI

    42. Association of Disease-free survival (DFS) with DNA-TG for R3-TEAM [5 year estimates from the time of randomisation will be measured but adequate follow-up for these estimates will be ensured: at least 2-year follow-up]

      Disease-free survival (DFS) - as defined above associated with DNA-TG.

    43. Cumulative incidence of Sinusoidal Obstruction Syndrome (SOS) and Nodular Regenerative Hyperplasia (NRH) for R3-TEAM [Cumulative incidence of SOS/NRH estimated at the end of follow-up.]

      Time from randomisation until diagnosis of SOS or NRH as defined in the protocol or end of follow-up.

    44. Cumulative incidence of Osteonecrosis for R3-TEAM [Cumulative incidence of osteonecrosis estimated at the end of follow-up.]

      Time from randomisation until diagnosis of osteonecrosis as defined in the protocol or end of follow-up.

    45. Event-free survival (EFS) for ALLTogether1 DS [From the start of Blinatumomab, 5 year estimate will be measured but adequate follow-up for this estimate will be ensured: at least 5 years follow-up.]

      Event-free survival as defined in the protocol, from the start of Blinatumomab until death from any cause, relapse, second malignancy, protocol therapy failure (MRD>1% after 2 cycles blinatumomab and Augmented BFM consolidation) or end of follow-up.

    46. Incidence of Blinatumomab refractory disease for ALLTogether1 DS [From the start of Blinatumomab until end of 2nd cycle of Blinatumomab (each cycle is 4 weeks followed by a 2-week treatment free period)]

      Incidence of progressive disease under Blinatumomab treatment or MRD ≥1% at the end of the 2nd cycle of Blinatumomab.

    47. Incidence of Protocol Therapy Failure for ALLTogether1 DS [From the start of Blinatumomab until the end of Consolidation 1 (85-140 days: 15-70 days of Blinatumomab therapy + 70 days of Consolidation 1)]

      Incidence of patients who have MRD ≥1 % post 2 cycles of Blinatumomab followed by Augmented BFM consolidation, corresponding to original protocol day 99, or unchanged/increasing MRD during Augmented BFM consolidation corresponding to original protocol day 57.

    Other Outcome Measures

    1. 6-mercaptopurine and Methotrexate metabolite pharmacokinetics (i.e. Ery-TGN/MeMP/MTXpg) for R3-TEAM [From start of Maintenance therapy until the end of Maintenance therapy (protocol week 38 until protocol week 108)]

      Measurements of metabolites Ery-TGN/MeMP/MTXpg during Maintenance therapy.

    2. Abnormal liver function parameters (including hypoglycemia) for R3-TEAM [From start of Maintenance therapy until the end of Maintenance therapy (protocol week 38 until protocol week 108)]

      Liver function parameters including hypoglycemia during Maintenance therapy

    Eligibility Criteria

    Criteria

    Ages Eligible for Study:
    0 Years to 45 Years
    Sexes Eligible for Study:
    All
    Accepts Healthy Volunteers:
    No
    Inclusion Criteria:
    • Patients newly diagnosed with T-lymphoblastic (T-cell) or B-lymphoblastic precursor (BCP) leukaemia (ALL) according to the WHO-classification of Tumours of Haematopoetic and Lymphoid Tissues (Revised 4th edition 2017) and with a diagnosis confirmed by an accredited laboratory at a participating paediatric oncology or adult haematology centre.

    • Age ≥ 0 days and < 46 years (one day before 46th birthday) at the time of diagnosis with the exception of infants with KMT2A-rearranged (KMT2A-r) BCP ALL.

    • Informed consent signed by the patient and/or parents/legal guardians according to country-specific age-related guidelines (http://www.ema.europa.eu/docs/en_GB/document_library/Other/2015/12/WC500199234.pdf ).

    • The ALL diagnosis should be confirmed by an accredited laboratory at a participating paediatric oncology or adult haematology centre.

    • The patient should be diagnosed and treated at a participating paediatric oncology or adult haematology centre in the participating countries.

    • The patient should be a resident in one of the participating countries on a permanent basis or should intend to settle in a participating country, for instance by an application for asylum. Patients who are visiting the country as tourists should not be included. However, returning expatriots with primary diagnosis abroad may be included if no treatment has been administered and the diagnostic procedures are repeated at a participating centre.

    • All women of childbearing potential (WOCBP) have to have a negative pregnancy test within 2 weeks prior to the start of treatment.

    • For each intervention/randomisation an additional set of inclusion-criteria is provided.

    Exclusion Criteria:
    • Age < 365 days and KMT2A-rearranged (KMT2A-r) BCP-ALL (documented precence of a KMT2A-split by FISH and/or a KMT2A fusion transcript).

    • Age >45 years at diagnosis.

    • Patients with a previous malignant diagnosis (ALL as a second malignant neoplasm - SMN).

    • Relapse of ALL.

    • Patients with mature B-ALL (as defined by Surface Ig positivity or documented presence of one of the t(8;14), t(2;8), t(8;22) translocations and breakpoint as in B-ALL).

    • Patients with Ph-positive ALL (documented presence of t(9;22)(q34;q11) and/or of the BCR/ABL fusion transcript). These patients will be transferred to an adequate trial for t(9;22) if available.

    • ALL prone syndromes (e.g. Li-Fraumeni syndrome, germline ETV6 mutation), except for Down syndrome. Exploration for such ALL prone syndromes is not mandatory.

    • Treatment with systemic corticosteroids (>10mg/m2/day) for more than one week and/or other chemotherapeutic agents in a 4-week interval prior to diagnosis (pre-treatment).

    • Pre-existing contraindications to any treatment according to the ALLTogether protocol (constitutional or acquired disease prior to the diagnosis of ALL preventing adequate treatment).

    • Any other disease or condition, as determined by the investigator, which could interfere with the participation in the study according to the study protocol, or with the ability of the patients to cooperate and comply with the study procedures.

    • Women of childbearing potential who are pregnant at the time of diagnosis.

    • Women of childbearing potential and fertile men who are sexually active and are unwilling to use adequate contraception during therapy. Efficient birth control is required.

    • Female patients, who are breast-feeding.

    • Essential data missing from the registration of characteristics at diagnosis (in consultation with the protocol chair).

    • For each intervention/randomisation an additional set of exclusion-criteria is provided.

    Contacts and Locations

    Locations

    Site City State Country Postal Code
    1 L'hôpital Universitaire des enfants Reine Fabiola (Huderf) Brussels Belgium 1020
    2 University Hospital Brussels Brussels Belgium 1090
    3 Cliniques Universitaires Saint-Luc (UCL) Brussels Belgium 1200
    4 University Hospital Antwerp Edegem Belgium 2650
    5 University Hospital Ghent Ghent Belgium 9000
    6 University Hospital Leuven, Dept of Haematology Leuven Belgium 3000
    7 University Hospital Leuven, Dept of Paediatrics Leuven Belgium 3000
    8 CHC MontLégia, Boulevard Patience et Beaujonc 2 Liège Belgium 4000
    9 CHR de la Citadelle Liège Belgium 4000
    10 Aalborg University Hospital, Dept of Paediatrics Aalborg Denmark 9000
    11 Aarhus University Hospital Aarhus Denmark 8000
    12 Aarhus University Hospital, Child and Adolescent Health Aarhus Denmark 8200
    13 Rigshospitalet, Dept of Haematology Copenhagen Denmark 2100
    14 Rigshospitalet, Dept of Paediatrics Copenhagen Denmark 2100
    15 Odense University Hospital, Dept of Paediatrics Odense Denmark 5000
    16 North Estonia Medical Centre, Dept of Haematology Tallinn Estonia 13419
    17 Tallinn Children´s Hospital, Dept of Paediatrics Tallinn Estonia 13419
    18 Tartu University Hospital, Dept of Paediatrics Tartu Estonia 50406
    19 Tartu University Hospital Tartu Estonia 50406
    20 Helsinki University Hospital, Dept of Haematology Helsinki Finland 00029
    21 Helsinki University Hospital, Dept of Paediatrics Helsinki Finland 00029
    22 Kuopio University Hospital, Dept of Haematology Kuopio Finland 70029
    23 Kuopio University Hospital, Dept of Paediatrics Kuopio Finland 70029
    24 Oulu University Hospital, Dept of Haematology, Dept of Medicine Oulu Finland 90029
    25 Oulu University Hospital, Dept of Paediatrics Oulu Finland 90029
    26 Tampere University Hospital, Dept of Haematology Tampere Finland 33521
    27 Tampere University Hospital, Dept of Paediatrics Tampere Finland 33521
    28 Turku University Hospital, Clinical Haematology and Stem Cell Transplantation Unit Turku Finland 20520
    29 Turku University Hospital, Dept of Paediatrics Turku Finland 20520
    30 Evangelisches Krankenhaus Bielefeld Bielefeld Germany 33617
    31 Universitätsklinikum Bonn Bonn Germany 53113
    32 Klinikum Bremen Mitte Bremen Germany 28177
    33 Universitätsklinikum Hamburg-Eppendorf Hamburg Germany 20246
    34 HELIOS Klinikum Krefeld Krefeld Germany 47805
    35 Universitätsmedizin Mainz Mainz Germany 55131
    36 Landspitali University Hospital, Children's Hospital Reykjavík Iceland 101
    37 Our Lady's Children's Hospital Dublin Ireland
    38 Children's Hospital, Affiliate of Vilnius University Hospital Santaros Klinikos Vilnius Lithuania 08661
    39 Vilnius University Hospital Santaros Klinikos Vilnius Lithuania 08661
    40 Princess Máxima Center for Pediatric Oncology Utrecht Netherlands 3584
    41 Haukeland University Hospital, Dept of Haematology Bergen Norway 5021
    42 Haukeland University Hospital, Dept of Paediatrics Bergen Norway 5021
    43 Oslo University Hospital, Dept of Haematology Oslo Norway 0372
    44 Oslo University Hospital, Dept of paediatric haemato- and oncology Oslo Norway 0424
    45 Stavanger University Hospital, Dept of Haematology Stavanger Norway 4011
    46 University Hospital North Norway, Dept of Haematology Tromsø Norway 9019
    47 University Hospital of North Norway, Dept of Paediatrics Tromsø Norway 9038
    48 St. Olavs University Hospital, Dept of Paediatrics Trondheim Norway 7006
    49 St. Olavs University Hospital, Dept of Haematology Trondheim Norway 7030
    50 Centro Hospitalar e Universitário de Coimbra, EPE - Hospital Pediátrico de Coimbra Coimbra Portugal 3000-602
    51 Instituto Português de Oncologia de Lisboa Francisco Gentil, EPE Lisboa Portugal 1099-023
    52 Instituto Português de Oncologia do Porto Francisco Gentil, EPE Porto Portugal 4200-072
    53 Sahlgrenska University Hospital, Section for Haematology and coagulation Gothenburg Sweden 41345
    54 Sahlgrenska University Hospital, Dept of Paediatric Haematology and Oncology Gothenburg Sweden 41685
    55 Linköping University Hospital, Dept of Haematology Linköping Sweden 58185
    56 Linköping University Hospital, Dept of Paediatrics Linköping Sweden 58185
    57 Skåne University Hospital, Dept of Haematology Lund Sweden 22185
    58 Skåne University Hospital, Dept of Paediatrics Lund Sweden 22185
    59 Karolinska University Hospital, Dept of Paediatric Oncology and Haematology Stockholm Sweden 17176
    60 Karolinska University Hospital, Patient area Haematology Stockholm Sweden 17176
    61 Norrland University Hospital, Dept of Haematology Umeå Sweden 90185
    62 Norrland University Hospital, Dept of Paediatrics Umeå Sweden 90185
    63 Uppsala University Hospital, Dept of Haematology Uppsala Sweden 75185
    64 Uppsala University Hospital, Dept of Paediatric Haematology and Oncology Uppsala Sweden 75185
    65 Örebro University Hospital, Section for Haematology Örebro Sweden 70185
    66 Aberdeen Royal Infirmary, Aberdeen Aberdeen United Kingdom AB25 2ZN
    67 Royal Aberdeen Children's Hospital, Aberdeen Aberdeen United Kingdom
    68 Royal Belfast Hospital for Sick Children, Belfast Belfast United Kingdom BT12 6BA
    69 Belfast City Hospital, Belfast Belfast United Kingdom BT9 7AB
    70 The Queen Elizabeth Hospital, Birmingham Birmingham United Kingdom B15 2TH
    71 Birmingham Children's Hospital, Birmingham Birmingham United Kingdom B4 6NH
    72 Bristol Royal Hospital for Children / Bristol Haematology and Oncology Centre Bristol United Kingdom BS2 8BJ
    73 Addenbrooke's Hospital, Cambridge Cambridge United Kingdom CB2 0QQ
    74 Noah's Ark Children's Hospital for Wales, Cardiff Cardiff United Kingdom CF14 4XW
    75 University Hospital of Wales, Cardiff Cardiff United Kingdom
    76 Ninewells Hospital, Dundee Dundee United Kingdom DD1 9SY
    77 Western General Hospital, Edinburgh Edinburgh United Kingdom EH2 2XU
    78 Royal Hospital for Sick Children, Edinburgh Edinburgh United Kingdom EH9 1LF
    79 Beatson West of Scotland Cancer Centre, Glasgow Glasgow United Kingdom G12 0YN
    80 Royal Hospital for Children, Glasgow Glasgow United Kingdom G51 4TF
    81 Leeds General Infirmary, Leeds Leeds United Kingdom LS1 3EX
    82 St. James's University Hospital, Leeds Leeds United Kingdom LS9 7TF
    83 Leicester Royal Infirmary, Leicester Leicester United Kingdom LE1 5WW
    84 Alder Hey Children's Hospital, Liverpool Liverpool United Kingdom L12 2AP
    85 Royal Liverpool University Hospital, Liverpool Liverpool United Kingdom
    86 University College London Hospital, London London United Kingdom NW1 2BU
    87 Great Ormond Street Hospital for Children, London London United Kingdom WC1N 3JH
    88 Royal Manchester Children's Hospital, Manchester Manchester United Kingdom M13 9WL
    89 Christie Hospital, Manchester Manchester United Kingdom M20 4BX
    90 Royal Victoria Infirmary, Newcastle Newcastle United Kingdom NE1 4LP
    91 Freeman Hospital, Newcastle Newcastle United Kingdom NE7 7DN
    92 Nottingham City Hospital, Nottingham Nottingham United Kingdom NG5 1PB
    93 Queen's Medical Centre, Nottingham Nottingham United Kingdom NG7 2UH
    94 Churchill Hospital, Oxford Oxford United Kingdom OX3 7LE
    95 John Radcliffe Hospital, Oxford Oxford United Kingdom OX3 9DU
    96 Derriford Hospital, Plymouth Plymouth United Kingdom PL6 8DH
    97 Royal Hallamshire Hospital, Sheffield Sheffield United Kingdom S10 2JF
    98 Sheffield Children's Hospital, Sheffield Sheffield United Kingdom S10 2TH
    99 Southampton General Hospital, Southampton Southampton United Kingdom SO16 6YD
    100 Royal Stoke University Hospital, Stoke Stoke United Kingdom ST4 6QG
    101 Royal Marsden Hospital, Sutton Sutton United Kingdom SM2 5PT

    Sponsors and Collaborators

    • Mats Heyman
    • The Swedish Research Council
    • The Swedish Childhood Cancer Foundation
    • Pfizer
    • Servier
    • NordForsk
    • Aamu Pediatric Cancer Foundation
    • Gesellschaft fur Padiatrische Onkologie und Hamatologie - Germany
    • Clinical Trial Center North (CTC North GmbH & Co. KG)
    • Belgium Health Care Knowledge Centre
    • Karolinska Institutet
    • Cancer Research UK
    • Fundação Rui Osório de Castro
    • Acreditar - Associação de Pais e Amigos das Crianças com Cancro
    • Grupo Português De Leucemias Pediátricas
    • Amgen
    • Nova Laboratories Limited
    • Danish Child Cancer Foundation
    • Danish Cancer Society
    • The Novo Nordic Foundation

    Investigators

    • Study Chair: Mats Heyman, MD, PhD, Karolinska University Hospital

    Study Documents (Full-Text)

    None provided.

    More Information

    Publications

    Responsible Party:
    Mats Heyman, MD, Associate Professor, Karolinska University Hospital
    ClinicalTrials.gov Identifier:
    NCT04307576
    Other Study ID Numbers:
    • ALLTogether1
    First Posted:
    Mar 13, 2020
    Last Update Posted:
    Aug 19, 2022
    Last Verified:
    Aug 1, 2022
    Individual Participant Data (IPD) Sharing Statement:
    Yes
    Plan to Share IPD:
    Yes
    Studies a U.S. FDA-regulated Drug Product:
    Yes
    Studies a U.S. FDA-regulated Device Product:
    No
    Product Manufactured in and Exported from the U.S.:
    Yes
    Keywords provided by Mats Heyman, MD, Associate Professor, Karolinska University Hospital
    Additional relevant MeSH terms:

    Study Results

    No Results Posted as of Aug 19, 2022