Tolerability and Efficacy of Midostaurin to 10-day Decitabine in Unfit Adult AML and High Risk MDS Patients (HO155)

A Randomized Phase II Multicenter Study to Assess the Tolerability and Efficacy of the Addition of Midostaurin to 10-day Decitabine Treatment in Unfit Adult Acute Myeloid Leukemia and High Risk Myelodysplasia Patients

The aim of this study is to investigate how safe and effective the addition of the new medicine midostaurin to decitabine is for the treatment of unfit acute myeloid leukemia (AML) and high-risk myelodysplasia (MDS) patients. Patients who are ineligible for intensive chemotherapy because of accompanying diseases may opt for gentler treatment. This does not produce a cure but serves to allow the quality of life to be acceptable for as long as possible. Decitabine is an example of a gentler treatment. It is effective against leukemia and has fewer side effects than intensive chemotherapy. Given in courses of 5 successive days, decitabine is registered for the treatment of AML. There is scientific research to suggest that decitabine is more effective and generally well tolerated when given in courses of 10 successive days. Therefore, treatment with 10-day courses of decitabine is the standard treatment in this scientific research. The aim is to investigate whether this standard treatment can be improved by adding a new product, midostaurin. Midostaurin is a medicine that is directed against a specific protein on leukaemia cells (FLT3).

Study Overview

Status

Active, not recruiting

Conditions

Detailed Description

This trial aims to develop effective treatments for unfit (i.e. Hematopoietic cell transplantation co-morbidity index (HCT-CI) ≥ 3) in adult (≥ 18 yrs) AML patients, for whom current treatment strategies are highly unsatisfactory. Therefore new treatment modalities are introduced and evaluated in multiple parallel randomized phase II studies that will be conducted within the frame of a master protocol. The scheme of this new design consists of one arm with one of the currently considered best available treatments for unfit AML patients (i.e. 10-day decitabine). After a maximum of 3 10-day courses, or less in case of good response, treatment will be continued with 5-day decitabine courses. This treatment will be compared to investigational treatments in combination with decitabine.

The competitor of the 10-day decitabine schedule will be 10-day decitabine combined (sequential) with the tyrosine kinase inhibitor midostaurin (independent of the presence of FLT3 mutations). The rationale for midostaurin is: 1) single agent midostaurin has shown efficacy in both FLT3 wild type and mutant AML; 2) it has shown efficacy in a phase III randomized controlled trial when combined with intensive chemotherapy in FLT3-mutated AML (RATIFY study); 3) midostaurin has been successfully combined with hypomethylating agents (azacitidine and decitabine) and improved the response compared with historical response rates of these drugs, suggesting at least additive affects of midostaurin with hypomethylating agents.

Study Type

Interventional

Enrollment (Actual)

140

Phase

  • Phase 2

Contacts and Locations

This section provides the contact details for those conducting the study, and information on where this study is being conducted.

Study Locations

      • Antwerpen, Belgium
        • BE-Antwerpen-ZNASTUIVENBERG
      • Haine-Saint-Paul, Belgium
        • BE-Haine-Saint-Paul-JOLIMONT
      • Roeselare, Belgium
        • BE-Roeselare-AZDELTA
      • Magdeburg, Germany
        • DE-Magdeburg-OVGU
      • Amersfoort, Netherlands
        • NL-Amersfoort-MEANDERMC
      • Amsterdam, Netherlands
        • NL-Amsterdam-OLVG
      • Amsterdam, Netherlands
        • NL-Amsterdam-VUMC
      • Arnhem, Netherlands
        • NL-Arnhem-RIJNSTATE
      • Breda, Netherlands
        • NL-Breda-AMPHIA
      • Delft, Netherlands
        • NL-Delft-RDGG
      • Den Bosch, Netherlands
        • NL-Den Bosch-JBZ
      • Den Haag, Netherlands
        • NL-Den Haag-HAGA
      • Doetinchem, Netherlands
        • NL-Doetinchem-SLINGELAND
      • Dordrecht, Netherlands
        • NL-Dordrecht-ASZ
      • Ede, Netherlands
        • NL-Ede-ZGV
      • Eindhoven, Netherlands
        • NL-Eindhoven-CATHARINA
      • Eindhoven, Netherlands
        • NL-Eindhoven-MAXIMAMC
      • Enschede, Netherlands
        • NL-Enschede-MST
      • Groningen, Netherlands
        • NL-Groningen-UMCG
      • Leeuwarden, Netherlands
        • NL-Leeuwarden-MCL
      • Maastricht, Netherlands
        • NL-Maastricht-MUMC
      • Nieuwegein, Netherlands
        • NL-Nieuwegein-ANTONIUS
      • Nijmegen, Netherlands
        • NL-Nijmegen-CWZ
      • Nijmegen, Netherlands
        • NL-Nijmegen-RADBOUDUMC
      • Rotterdam, Netherlands
        • NL-Rotterdam-ERASMUSMC
      • Utrecht, Netherlands
        • NL-Utrecht-UMCUTRECHT
      • Zwolle, Netherlands
        • NL-Zwolle-ISALA
      • Aarau, Switzerland
        • CH-Aarau-KSA
      • Basel, Switzerland
        • CH-Basel-USB
      • Bellinzona, Switzerland
        • CH-Bellinzona-IOSI
      • Bern, Switzerland
        • CH-Bern-INSEL
      • Fribourg, Switzerland
        • CH-Fribourg-HFR
      • Geneve, Switzerland
        • CH-Geneve (14)-HCUGE
      • Lausanne, Switzerland
        • CH-Lausanne-CHUV
      • Luzern, Switzerland
        • CH-Luzern-LUKS
      • Saint Gallen, Switzerland
        • CH-St. Gallen-KSSG
      • Zürich, Switzerland
        • CH-Zürich-USZ

Participation Criteria

Researchers look for people who fit a certain description, called eligibility criteria. Some examples of these criteria are a person's general health condition or prior treatments.

Eligibility Criteria

Ages Eligible for Study

18 years to 100 years (Adult, Older Adult)

Accepts Healthy Volunteers

No

Genders Eligible for Study

All

Description

Inclusion Criteria:

  • Patients with:

    • a diagnosis of AML and related precursor neoplasms according to WHO 2016 classification (excluding acute promyelocytic leukemia) including secondary AML (after an antecedent hematological disease (e.g. MDS) and therapy-related AML, or
    • a diagnosis of myelodysplastic syndrome with excess of blasts (MDS) and International Prognostic Score System (IPSS) > 4.5
  • Patients 18 years and older.
  • Patients NOT eligible for standard chemotherapy, defined as hematopoietic cell transplantation comorbidity index (HCT-CI) ≥ 3.

or Patients NOT eligible for standard chemotherapy for other reasons (wish of patient).

  • White blood cell (WBC) ≤ 30 x109/L (prior hydroxyurea allowed for a maximum of 5 days, stop 2 days before start decitabine treatment)
  • Adequate renal and hepatic functions unless clearly disease related as indicated by the following laboratory values:

    • Serum creatinine ≤ 221.7 µmol/L (≤ 2.5 mg/dL ), unless considered AML-related
    • Serum bilirubin ≤ 2.5 x upper limit of normal (ULN), unless considered AML-related or due to Gilbert's syndrome
    • Alanine transaminase (ALT) ≤ 2.5 x ULN, unless considered AML-related
  • WHO performance status 0, 1 or 2.
  • Patient is willing and able to use adequate contraception during and until 5 months after the last protocol treatment.
  • Written informed consent.
  • Patient is capable of giving informed consent.

Exclusion Criteria:

  • Acute promyelocytic leukemia.
  • Acute leukemia's of ambiguous lineage according to WHO 2016
  • Patient has symptomatic central nervous system (CNS) leukemia (NO routinely lumbar puncture required to investigate CNS involvement)
  • Blast crisis of chronic myeloid leukemia.
  • Diagnosis of any previous or concomitant malignancy is an exclusion criterion:
  • except when the patient completed successfully treatment (chemotherapy and/or surgery and/or radiotherapy) with curative intent for this malignancy at least 6 months prior to randomization. OR
  • except for basal and squamous cell carcinoma of the skin or in situ carcinoma of the cervix
  • Patients previously treated for AML (any antileukemic therapy including investigational agents), a short treatment period ( ≤ 5 days) with Hydroxyurea is allowed
  • Current concomitant chemotherapy, radiation therapy, or immunotherapy; other than hydroxyurea
  • Concurrent severe and/or uncontrolled medical condition (e.g. uncontrolled diabetes, infection, hypertension, pulmonary disease etc.)
  • Cardiac dysfunction as defined by:

    • Myocardial infarction within the last 3 months of study entry, or
    • Reduced left ventricular function with an ejection fraction < 40% as measured by MUGA scan or echocardiogram or
    • Unstable angina or
    • New York Heart Association grade IV congestive heart failure or
    • Unstable cardiac arrhythmias.
  • History of stroke or intracranial hemorrhage within 6 months prior to randomization.
  • Patient has a history of human immunodeficiency virus or active infection with Hepatitis C or B.
  • Patients known to be pregnant
  • Patients with a history of non-compliance to medical regimens or who are considered unreliable with respect to compliance.
  • Patients with any serious concomitant medical condition which could, in the opinion of the investigator, compromise participation in the study.
  • Patients who have senile dementia, mental impairment or any other psychiatric disorder that prohibits the patient from understanding and giving informed consent.
  • Any psychological, familial, sociological or geographical condition potentially hampering compliance with the study protocol and follow-up schedule

Study Plan

This section provides details of the study plan, including how the study is designed and what the study is measuring.

How is the study designed?

Design Details

  • Primary Purpose: Treatment
  • Allocation: Randomized
  • Interventional Model: Parallel Assignment
  • Masking: None (Open Label)

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
Active Comparator: Arm A: Decitabine

Cycles 1-3: Decitabine 10-day; depending on day +28 bone marrow (BM) blasts after the previous cycle, next cycle consists of either 5-day (BM blasts < 5%) or 10-day (BM blasts ≥5%) decitabine. Cycles 4 and beyond: 5-day decitabine (in cycles of 4-8 weeks); continuation of these cycles until progression.

Dosage for Decitabine 20 mg/m2 i.v.

Decitabine dosage 20mg/m2 i.v.
Other Names:
  • Dacogen
Experimental: Arm B: Decitabine and Midostaurin

Cycle 1:Decitabine; 10-day schedule (start day +1) + midostaurin (start day +11). Midostaurin is given until 2 days before start next cycle of decitabine. Cycles 2-3: Decitabine 5 or 10-day schedule; depending on day +28 bone marrow blasts of the previous cycle, next cycle consist of either 5-day (BM blasts < 5%) or 10-day (BM blasts ≥5%) decitabine + midostaurin (daily, starting the day after the last dose of decitabine (i.e. day +6 or +11). Midostaurin is given until 2 days before start next cycle. Cycles 4 and beyond: 5-day decitabine (in cycles of 4-8 weeks) followed by midostaurin starting at day +6 until two days before start of next cycle of decitabine; continuation of these cycles until progression. Midostaurin is given until 2 days before start next cycle of decitabine.

Dosage for Decitabine 20 mg/m2 i.v.

Dosage for Midostaurin 50 mg b.i.d.

Decitabine dosage 20mg/m2 i.v.
Other Names:
  • Dacogen
Midostaurin 50 mg b.i.d.
Other Names:
  • Rydapt

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Cumulative Complete Remission (CR) / CR with incomplete blood count (CRi) rate
Time Frame: 4-5 months
Cumulative CR/CRi rate during 3 cycles
4-5 months

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Safety and tolerability of midostaurin determined by the type, frequency, severity and relationship of adverse events to study treatment
Time Frame: 5 years
Safety and tolerability of midostaurin added to 10-day decitabine treatment for AML (type, frequency, severity and relationship of adverse events to study treatment).
5 years
Efficacy profile
Time Frame: 4-9 months
The response rate after first three cycles together with the best response during three cycles and after 9 months will determinine the efficacy profile.
4-9 months
Event free survival (EFS)
Time Frame: 5 years
The time from registration to induction failure, death or relapse whichever occurs first).
5 years
Overall survival (OS)
Time Frame: 5 years
The time from the date of randomization to the date of death, whatever the cause. Patients still alive at the date last contact will be censored.
5 years
Hospital stay duration
Time Frame: 4-5 months
Days of staying in hospital during 3 cycles.
4-5 months
Transfusion need
Time Frame: 4-5 months
Number of participants with transfusion needs during 3 cycles.
4-5 months
Prognostic value of MRD
Time Frame: 9 months and at relapse
Assessment of the prognostic value of Minimal Residual Disease (MRD) by flowcytometry or PCR
9 months and at relapse
Predictive value of gene mutations
Time Frame: 5 years
Assessment of the predictive value of gene mutations by exploratory analysis
5 years
Prognostic value of baseline physical conditions as measured by the short physical performance battery
Time Frame: 5 years
Assessment of the prognostic value of baseline physical and functional conditions using a comprehensive geriatric assessment tool, short physical performance battery (SPPB), on treatment outcome. Total scores (range 0 to 12) will be used to determine physical performance.
5 years
Prognostic value of baseline functional conditions as measured by the activities of daily living
Time Frame: 5 years
Assessment of the prognostic value of baseline functional conditions using a comprehensive geriatric assessment tool, activities of daily living (ADL), on treatment outcome. Total points (range 0 to 6) will be used to determine functional condition.
5 years

Collaborators and Investigators

This is where you will find people and organizations involved with this study.

Investigators

  • Principal Investigator: Gerwin Huls, Prof, UMCG / HOVON

Publications and helpful links

The person responsible for entering information about the study voluntarily provides these publications. These may be about anything related to the study.

Helpful Links

Study record dates

These dates track the progress of study record and summary results submissions to ClinicalTrials.gov. Study records and reported results are reviewed by the National Library of Medicine (NLM) to make sure they meet specific quality control standards before being posted on the public website.

Study Major Dates

Study Start (Actual)

December 5, 2019

Primary Completion (Actual)

November 15, 2021

Study Completion (Anticipated)

November 1, 2026

Study Registration Dates

First Submitted

March 25, 2019

First Submitted That Met QC Criteria

September 18, 2019

First Posted (Actual)

September 20, 2019

Study Record Updates

Last Update Posted (Estimate)

January 6, 2023

Last Update Submitted That Met QC Criteria

January 5, 2023

Last Verified

January 1, 2023

More Information

Terms related to this study

Other Study ID Numbers

  • HOVON 155 AML
  • 2018-000047-31 (EudraCT Number)
  • 2018-674 (Other Identifier: METc UMCG)
  • NL64632.042.18 (Other Identifier: CCMO)

Plan for Individual participant data (IPD)

Plan to Share Individual Participant Data (IPD)?

No

Drug and device information, study documents

Studies a U.S. FDA-regulated drug product

No

Studies a U.S. FDA-regulated device product

No

This information was retrieved directly from the website clinicaltrials.gov without any changes. If you have any requests to change, remove or update your study details, please contact register@clinicaltrials.gov. As soon as a change is implemented on clinicaltrials.gov, this will be updated automatically on our website as well.

Clinical Trials on AML/MDS

Clinical Trials on Decitabine

3
Subscribe