Phase III Study of Induction and Consolidation Chemotherapy With Venetoclax in Patients With Newly Diagnosed AML or MDS-EB-2

June 6, 2023 updated by: Prof. Dr. Hartmut Doehner, University of Ulm

A Randomized, Placebo-Controlled Phase III Study of Induction and Consolidation Chemotherapy With Venetoclax in Adult Patients With Newly Diagnosed Acute Myeloid Leukemia or Myelodysplastic Syndrome With Excess Blasts-2

A Randomized, Placebo-Controlled Phase III Study of Induction and Consolidation Chemotherapy With Venetoclax in Adult Patients With Newly Diagnosed Acute Myeloid Leukemia or Myelodysplastic Syndrome With Excess Blasts-2

Study Overview

Detailed Description

Prospective, multicenter, double-blind, randomized, placebo-controlled phase 3 clinical study. The randomized phase of the study will be preceded by a feasibility run-in dose-escalation phase in patients with AML in which the venetoclax dose for the phase 3 part will be established.

After the feasibility run-in phase, eligible patients will be randomized to intensive chemotherapy with venetoclax or placebo. Patients will receive two cycles of induction chemotherapy; patients achieving CR or CRi after two cycles will continue with consolidation treatment according to initial randomization, and according to Cooperative Group-specific consolidation regimens or investigator choice. Patients achieving morphologic leukemia-free state (MLFS) only, may also continue consolidation treatment on protocol. Assignment to either allogeneic hematopoietic cell transplantation (HCT), conventional chemotherapy or autologous HCT will be done according to institutional standards, and based on (prognostic) disease characteristics, individual patient assessment, and established comorbidity risk scores (e.g., HCT-CI score).

Study Type

Interventional

Enrollment (Estimated)

650

Phase

  • Phase 3

Contacts and Locations

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

Study Contact

Study Locations

      • Berlin, Germany, 12200
        • Recruiting
        • Charité Berlin - Campus Benjamin Franklin
        • Contact:
          • Jan Kroenke, Porf.
      • Berlin, Germany, 10117
        • Recruiting
        • Charité Berlin - Campus Mitte
        • Contact:
          • Jan Kroenke, Prof.
      • Berlin, Germany
        • Recruiting
        • Charité Berlin - Campus Virchow Klinikum
        • Contact:
          • Jan Kroenke, Prof.
      • Bochum, Germany, 44892
        • Recruiting
        • Knappschaftskrankenhaus Bochum-Langendreer
        • Contact:
          • Roland Schroers, Prof.
      • Bonn, Germany, 53127
        • Recruiting
        • Uniklinikum Bonn
        • Contact:
          • Lino Teichmann, Dr.
      • Braunschweig, Germany, 38114
        • Recruiting
        • Staedtisches Klinikum Braunschweig
        • Contact:
          • Jürgen Krauter, Prof.
      • Hamburg, Germany, 20246
        • Recruiting
        • Universitätsklinikum Hamburg-Eppendorf
        • Contact:
          • Walter Fiedler, Prof.
      • Hannover, Germany, 30625
        • Recruiting
        • Medizinische Hochschule Hannover
        • Contact:
          • Michael Heuser, Prof.
      • Karlsruhe, Germany, 76133
        • Recruiting
        • Städtisches Klinikum Karlsruhe
        • Contact:
          • Mark Ringhoffer, Prof.
      • Stuttgart, Germany, 70176
        • Recruiting
        • Diakonie Klinikum Stuttgart
        • Contact:
          • Jochen Greiner, Prof
      • Tübingen, Germany, 72076
        • Recruiting
        • Uniklinikum Tübingen
        • Contact:
          • Claudia Lengerke, Prof. Dr.
      • Ulm, Germany, 89081
        • Recruiting
        • University Hospital Ulm
        • Contact:
          • Hartmut Döhner, Prof. Dr.
        • Contact:
          • Gaidzik Verena, PD Dr.

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 and older (Adult, Older Adult)

Accepts Healthy Volunteers

No

Description

Inclusion Criteria:

  1. Patients with newly diagnosed acute myeloid leukemia (AML), or myelodysplastic syndrome with excess blasts-2 (MDS-EB2) according to World Health Organization (WHO) classification.
  2. Age ≥ 18 years, no upper age limit.
  3. Patient considered eligible for intensive chemotherapy.
  4. Eastern Cooperative Oncology Group (ECOG) performance status ≤ 2 at randomization.
  5. Molecular analysis centrally performed in AMLSG and HOVON laboratories.
  6. Adequate renal function as evidenced by serum creatinine ≤ 2.0 × upper limit of norm (ULN) or creatinine clearance >40 mL/min based on the Cockcroft-Gault glomerular filtration rate (GFR).
  7. Adequate hepatic function as evidenced by:

    • Serum total bilirubin ≤ 2.5 × ULN unless considered due to Gilbert's disease, or leukemic involvement following approval by the Coordinating Investigator or Trial Coordinator
    • Aspartate aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphatase (ALP) ≤ 3.0 × ULN, unless considered due to leukemic involvement following approval by the Coordinating Investigator or Trial Coordinator
  8. No prior chemotherapy for AML except hydroxyurea for up to 14 days during the diagnostic screening phase for the control of peripheral leukemic blasts in patients with leukocytosis (e.g., white blood cell [WBC] counts > 25x109/L); patients may have had previous treatment with erythroid stimulating agents (ESA) or hypomethylating agents (HMAs) for an antecedent phase of MDS; ESA and HMAs have to be stopped at least four weeks before enrolment.
  9. Subjects must not have received a known strong or moderate CYP3A inducer 7 days before enrolment. Subjects must have no known medical conditions requiring chronic therapy with moderate or strong CYP3A inducers.
  10. Female patient must either:

    • Be of nonchildbearing potential:

      • Postmenopausal (defined as at least 1 year without any menses)
      • Documented surgically sterile or status posthysterectomy (at least 1 month prior to screening)
    • Or, if of childbearing potential (not surgically sterile (e.g. documented hysterectomy, bilateral oophorectomy, bilateral salpingectomy or congenital sterile) and not postmenopausal)

      • Not planning to become pregnant during the study and for 6 months after the final study drug administration
      • And have a negative urine or serum pregnancy test at screening
      • And, if heterosexually active, agree to consistently apply one highly effective* in combination to a barrier method for the duration of the study and for 6 months after the final study drug administration

        *Highly effective forms of birth control include

      • Consistent and correct usage of established hormonal contraceptives that inhibit ovulation (hormonal contraception is only a highly effective method of birth control, if a combined [estrogen and progestogen containing] hormonal contraception or a progestogen-only hormonal contraception - both associated with inhibition of ovulation - is used.
      • Established intrauterine device (IUD) or intrauterine system (IUS)
      • Bilateral tubal occlusion
      • Vasectomy - a vasectomy is highly effective contraception method provided the absence of sperm has been confirmed. If not, an additional highly effective method of contraception should be used.
      • Male is sterile due to a bilateral orchiectomy.
      • Sexual abstinence is considered a highly effective method only if defined as refraining from heterosexual activity during the entire period of risk associated with the study drug. The reliability of sexual abstinence needs to be evaluated in relation to the duration of the clinical study and the preferred and usual lifestyle of the patient.

        *List is not all inclusive. Prior to enrolment, the investigator is responsible for confirming patient will utilize highly effective forms of birth control in combination with a barrier method according to locally accepted standards during the protocol defined period.

      • Female patient must agree not to breastfeed starting at screening and throughout the study period, and for 2 months and 1 week after the final study drug administration.
      • Female patient must not donate ova starting at screening and throughout the study period, and for 6 months after the final study drug administration.
  11. Men must use a latex condom during any sexual contact with WOCBP, even if they have undergone a successful vasectomy and must agree to avoid to father a child (while on therapy and for 6 months after the final study drug administration). In addition, their female partners of childbearing potential have to use a highly effective method of birth control.
  12. Male patient must not donate sperm starting at screening and throughout the study period and for 6 months after the final study drug administration.
  13. Able to understand and willing to sign an informed consent form (ICF).

Exclusion Criteria:

  1. Acute promyelocytic leukemia (APL) with t(15;17)(q22;q12); PML-RARA; or one of the other pathognomonic variant chromosomal translocations / fusion genes.
  2. AML with BCR-ABL1; or myeloid blast crisis of CML.
  3. Patients with AML and activating FLT3 mutations who have access (including reimbursement) to treatment with a FLT3 inhibitor approved for first-line therapy of AML.
  4. Prior treatment of MDS with intensive chemotherapy or allogeneic hematopoietic cell transplantation (HCT) with a curative intent.
  5. Significant active cardiac disease within 6 months prior to the start of study treatment, including:

    • New York Heart Association (NYHA) class III or IV congestive heart failure;
    • Myocardial infarction;
    • Unstable angina and/or stroke;
    • Severe cardiac arrhythmias
    • Left ventricular ejection fraction (LVEF) <40% by ultrasound obtained within 28 days prior to the start of study treatment
  6. Severe obstructive or restrictive ventilation disorder.
  7. Co-administration of moderate/strong CYP inhibitors during the DLT observation period for subjects in the dose-finding part of the study.
  8. Clinical symptoms suggestive of active central nervous system (CNS) leukemia or known CNS leukemia. Evaluation of cerebrospinal fluid (CSF) during screening is only required, if there is a clinical suspicion of CNS involvement by leukemia during screening.
  9. Active infection, including hepatitis B or hepatitis C or Human Immunodeficiency Virus (HIV) infection, that is uncontrolled at randomization and may interfere with the study objectives or which could expose the subject to undue risk through the participation in the clinical trial; an infection controlled with an approved antibiotic/ antiviral/ antifungal treatment that is not a strong or moderate CYP3A inducer is allowed.
  10. Immediate life-threatening, severe complications of leukemia such as uncontrolled bleeding and/or disseminated intravascular coagulation.
  11. Conditions that limit the ingestion or gastrointestinal absorption of orally administered drugs.
  12. Patients with a currently active second malignancy. Patients are not considered to have a currently active malignancy, if they have completed therapy and are considered by their physician to be at < 30% risk of relapse within one year. However, subjects with the following history/concurrent conditions are allowed:

    • Basal or squamous cell carcinoma of the skin;
    • Carcinoma in situ of the cervix;
    • Carcinoma in situ of the breast;
    • Incidental histologic finding of prostate cancer.
  13. Receipt of live, attenuated vaccine within 30 days prior to the study inclusion (NOTE: subjects, if enrolled, should not receive live vaccine during the study and until 6 months after the therapy).
  14. Severe neurological or psychiatric disorder interfering with ability to give an informed consent.
  15. Known or suspected hypersensitivity to any of the chemotherapeutic agents used.
  16. No consent for registration, storage and processing of the individual disease characteristics and course as well as information of the family physician about study participation.
  17. No consent for biobanking of patient's biological specimens.
  18. Participation in other prospective studies with anti-leukemic and/or investigational agents.
  19. The patient is a pregnant or lactating woman, or plans to become pregnant during the study.

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: Quadruple

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
Experimental: 1
standard chemotherapy in combination with venetoclax
Induction cycle 1: Venetoclax will be administered orally QD, daily dose: 100mg/200mg/300mg, days 1-12 (after a 1/2/3 day ramp-up phase) (as determined during run-in phase) Induction cycle 2: Venetoclax will be administered orally QD, daily dose: 100mg/200mg/300mg, patients not in CR/CRi: days 1-12 (after a 1/2/3 day ramp-up phase) or patients in CR/CRi: days 1-14 (without ramp-up) (as determined during run-in phase) Consolidation phase: Venetoclax will be administered orally QD, daily dose: 100mg/200mg/300mg, days 1-14 (as determined during run-in phase)

Induction cycle 1: Patients will receive cytarabine 200 mg/m2 continuous IV (days 1-7) and daunorubicin 60 mg/m2 IV (days 1-3). Induction cycle 2: Patients ≤ 60 yrs will receive cytarabine 1000 mg/m2 BID (3h IV), days 1-6, and daunorubicin 60 mg/m2 IV (days 1-3). Patients >60 yrs will receive cytarabine 1000 mg/m2 BID (3h IV), days 1-6 without daunorubicin.

Consolidation chemotherapy option 1 consists of one cycle of mitoxantrone (10mg/m2 IV) and etoposide (100mg/m2 IV) (ME) days 1-5 (patients ≤60 yrs) or days 1-3 (patients >60 yrs). Consolidation chemotherapy option 2 consists of intermediate doses of cytarabine. Patients ≤60 yrs will receive up to 3 cycles of IDAC (single dose 1500 mg/m2 every 12 hours, days 1-3). Patients who are >60 yrs will receive up to 3 cycles of IDAC with single doses of 1000 mg/m2, every 12 hours, days 1-3. In patients >60 yrs less than 3 cycles of IDAC or dose-reduced IDAC (500 mg/m2 per single dose) may be given based on an individual risk assessment.

Busulfan/Cyclophosphamide will be administered as conditioning regime. Patients may proceed to autologous HCT when they have reached CR/CRi/MLFS after at least two cycles of chemotherapy and performance status permits continuation with high-dose chemotherapy. Venetoclax will not be added to or given after the conditioning regimen.
Generally, patients will proceed to allogeneic HCT upon completion of remission induction chemotherapy. It is however allowed, as per investigator's discretion, for a patient to receive 'bridging' consolidation chemotherapy in exceptional cases of delay towards transplantation. At baseline, HLA-compatible donor search must be initiated as soon as possible, first among siblings and second in the world donor bank for unrelated donors or cord blood. In order to avoid inappropriate delay in cases where no suitable sibling is present, high-resolution HLA typing should be performed immediately after registration, enabling a more rapid matched-unrelated donor search. In case no sibling or unrelated donor can be identified, haploidentical allogeneic HCT is allowed. Conditioning and GVHD prophylaxis will take place according to institutional guidelines. Patients who undergo allogeneic HCT will not receive venetoclax during conditioning, engraftment or after hematologic recovery.
Placebo Comparator: 2
standard chemotherapy in combination with placebo

Induction cycle 1: Patients will receive cytarabine 200 mg/m2 continuous IV (days 1-7) and daunorubicin 60 mg/m2 IV (days 1-3). Induction cycle 2: Patients ≤ 60 yrs will receive cytarabine 1000 mg/m2 BID (3h IV), days 1-6, and daunorubicin 60 mg/m2 IV (days 1-3). Patients >60 yrs will receive cytarabine 1000 mg/m2 BID (3h IV), days 1-6 without daunorubicin.

Consolidation chemotherapy option 1 consists of one cycle of mitoxantrone (10mg/m2 IV) and etoposide (100mg/m2 IV) (ME) days 1-5 (patients ≤60 yrs) or days 1-3 (patients >60 yrs). Consolidation chemotherapy option 2 consists of intermediate doses of cytarabine. Patients ≤60 yrs will receive up to 3 cycles of IDAC (single dose 1500 mg/m2 every 12 hours, days 1-3). Patients who are >60 yrs will receive up to 3 cycles of IDAC with single doses of 1000 mg/m2, every 12 hours, days 1-3. In patients >60 yrs less than 3 cycles of IDAC or dose-reduced IDAC (500 mg/m2 per single dose) may be given based on an individual risk assessment.

Busulfan/Cyclophosphamide will be administered as conditioning regime. Patients may proceed to autologous HCT when they have reached CR/CRi/MLFS after at least two cycles of chemotherapy and performance status permits continuation with high-dose chemotherapy. Venetoclax will not be added to or given after the conditioning regimen.
Generally, patients will proceed to allogeneic HCT upon completion of remission induction chemotherapy. It is however allowed, as per investigator's discretion, for a patient to receive 'bridging' consolidation chemotherapy in exceptional cases of delay towards transplantation. At baseline, HLA-compatible donor search must be initiated as soon as possible, first among siblings and second in the world donor bank for unrelated donors or cord blood. In order to avoid inappropriate delay in cases where no suitable sibling is present, high-resolution HLA typing should be performed immediately after registration, enabling a more rapid matched-unrelated donor search. In case no sibling or unrelated donor can be identified, haploidentical allogeneic HCT is allowed. Conditioning and GVHD prophylaxis will take place according to institutional guidelines. Patients who undergo allogeneic HCT will not receive venetoclax during conditioning, engraftment or after hematologic recovery.
Induction cycle 1: Placebo will be administered orally QD, daily dose: 100mg/200mg/300mg, days 1-12 (after a 1/2/3 day ramp-up phase) (as determined during run-in phase) Induction cycle 2: Placebo will be administered orally QD, daily dose: 100mg/200mg/300mg, patients not in CR/CRi: days 1-12 (after a 1/2/3 day ramp-up phase) or patients in CR/CRi: days 1-14 (without ramp-up) (as determined during run-in phase) Consolidation phase: Placebo will be administered orally QD, daily dose: 100mg/200mg/300mg, days 1-14 (as determined during run-in phase)

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Event Free Survival (EFS)
Time Frame: 6 months/16 months after inclusion of last patient
EFS in adult patients with newly diagnosed AML, defined as the time from randomization to treatment failure, death from any cause, or relapse after achieving CR or CRi, or start of new therapy due to confirmed molecular relapse whichever occurs first. Treatment failure is defined as not attaining CR or CRi after induction chemotherapy, and EFS event time for treatment failure is Day 1 of post-randomization
6 months/16 months after inclusion of last patient
Frequency of dose-limiting toxicities (DLTs) during the observation period (Primary safety endpoint during dose-finding phase)
Time Frame: after cycle 1 (maximal day 42)
Frequency of dose-limiting toxicities (DLTs) during the observation period (from start of cycle 1 up to a maximum of day 42, or until the start of cycle 2)
after cycle 1 (maximal day 42)

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Overall Survival (OS)
Time Frame: 6 months/16 months/28 months after inclusion of last patient
OS in patients with newly diagnosed AML
6 months/16 months/28 months after inclusion of last patient
CR/CRi rate
Time Frame: 2 months
Complete remission (CR/CRi) rate in newly diagnosed AML patients defined as the proportion of AML patients with CR/CRi after induction chemotherapy
2 months
CR rate
Time Frame: 2 months
Complete remission (CR) rates in newly diagnosed AML patients defined as the proportion of AML patients with CR after induction chemotherapy
2 months
Rates of complete remission without measurable residual disease (CRMRD-) after induction therapy
Time Frame: 2 months
Rates of complete remission without measurable residual disease (CRMRD-) after induction therapy, defined as the proportion of AML patients achieving CR/CRi with negativity for a genetic marker by RT-qPCR, and/or with negativity by multi-color flow cytometry, if studied pre-treatment
2 months
Relapse-free survival (RFS) in newly diagnosed AML patients
Time Frame: 16 months after inclusion of last patient
Relapse-free survival (RFS) in newly diagnosed AML patients, defined as the time from achievement of a remission (CR/CRi) following curative therapy (induction and consolidation), to relapse, or start of new therapy due to confirmed molecular relapse or death from any cause
16 months after inclusion of last patient
Cumulative incidence of relapse (CIR) in newly diagnosed AML patients
Time Frame: 16 months after inclusion of last patient
Cumulative incidence of relapse (CIR) in newly diagnosed AML patients
16 months after inclusion of last patient
Cumulative incidence of death (CID)
Time Frame: 16 months after inclusion of last patient
Cumulative incidence of death (CID) in newly diagnosed AML patients.
16 months after inclusion of last patient
EQ-5D-5L questionnaire of the EuroQoL (EQ) group, among AML patients
Time Frame: 6 months
Health status is measured in terms of five dimensions (5D); mobility, self-care, usual activities, pain/discomfort, and anxiety/depression. Each dimension is rated using a 5 level (5L) scale from 1 (minimum) to 5 (maximum). Higher score values mean a worse outcome.
6 months
European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC-QLQ-C30) among AML patients
Time Frame: 6 months
All sub scales and single items have values from 1 (minimum) to 100 (maximum) points. A higher point value represents a better condition in terms of the functional scales and quality of life. In the symptomal sub scales a higher point value represents a worse condition.
6 months
Patient Reported Outcome Measurement Information System (PROMIS) Cancer Fatigue short form among AML patients
Time Frame: 6 months
The 7-item PROMIS Cancer Fatigue Short Form assesses the frequency of fatigue in the past seven days (7). Table 2 presents a list of the items. Items are measured on a five point scale (1 = never; 5 = always) and summed, after reverse scoring item 7, with higher scores indicating greater fatigue (worse condition).
6 months

Other Outcome Measures

Outcome Measure
Measure Description
Time Frame
Rates of CR+CRi in newly diagnosed AML patients after induction 1
Time Frame: 1 month
Rates of CR+CRi in newly diagnosed AML patients defined as proportion of AML patients achieving CR/CRi after first course of induction
1 month
Rates of CR in newly diagnosed AML patients after induction 1
Time Frame: 1 month
Rates of CR in newly diagnosed AML patients defined as proportion of AML patients achieving CR after first course of induction
1 month
EFS in newly diagnosed AML patients across different patient subgroups
Time Frame: 6 months/16 months after inclusion of last patient
EFS in newly diagnosed AML patients across different groups are defined based on prognostic characteristics including age at registration , risk category according to ELN 2017 recommendations, as well as specific AML genotypes
6 months/16 months after inclusion of last patient
OS in newly diagnosed AML patients across different patient subgroups
Time Frame: 6 months/16 months/28 months after inclusion of last patient
OS in newly diagnosed AML patients across different patient subgroups, where the groups are defined based on prognostic characteristics including age at registration , risk category according to ELN 2017 recommendations, as well as specific AML genotypes
6 months/16 months/28 months after inclusion of last patient
CR/CRi rates in newly diagnosed AML patients across different patient subgroups
Time Frame: 1 month
CR/CRi rates in newly diagnosed AML patients across different patient subgroups, where the groups are defined based on prognostic characteristics including age at registration , risk category according to ELN 2017 recommendations, as well as specific AML genotypes
1 month
CR rates in newly diagnosed AML patients across different patient subgroups
Time Frame: 1 month
CR rates in newly diagnosed AML patients across different patient subgroups, where the groups are defined based on prognostic characteristics including age at registration , risk category according to ELN 2017 recommendations, as well as specific AML genotypes
1 month
RFS in newly diagnosed AML patients across different patient subgroups
Time Frame: 16 months after inclusion of last patient
RFS in newly diagnosed AML patients across different patient subgroups, where the groups are defined based on prognostic characteristics including age at registration , risk category according to ELN 2017 recommendations, as well as specific AML genotypes
16 months after inclusion of last patient
CIR in newly diagnosed AML patients across different patient subgroups
Time Frame: 16 months after inclusion of last patient
CIR in newly diagnosed AML patients across different patient subgroups, where the groups are defined based on prognostic characteristics including age at registration , risk category according to ELN 2017 recommendations, as well as specific AML genotypes
16 months after inclusion of last patient
CID in newly diagnosed AML patients across different patient subgroups
Time Frame: 16 months after inclusion of last patient
CID in newly diagnosed AML patients across different patient subgroups, where the groups are defined based on prognostic characteristics including age at registration , risk category according to ELN 2017 recommendations, as well as specific AML genotypes
16 months after inclusion of last patient
Frequency and severity of AE
Time Frame: 6 months
Frequency and severity of AE according to CTCAE version 5.0 in newly diagnosed AML patients and MDS-EB2 patients
6 months
Times to hematopoietic recovery
Time Frame: 6 months
Times to hematopoietic recovery (absolute neutrophil counts ≥0.5 and ≥1.0 x 109/L; platelets ≥50 and ≥100 x 109/L) after each chemotherapy treatment cycle, defined as the time from the start of the cycle until recovery among AML patients am MDS-EB2 patients
6 months
Frequency and severity of adverse events (AEs) (Secondary Safety endpoint during dose-finding phase)
Time Frame: 6 months
Frequency and severity of adverse events (AEs) according to Common Terminology Criteria for Adverse Events (CTCAE) version v5.0, see protocol Section 12.1 for definitions
6 months
EFS rates in MDS-EB2 patients
Time Frame: 6 months/16 months after inclusion of last patient
EFS rates in MDS-EB2 patients
6 months/16 months after inclusion of last patient
OS rates in MDS-EB2 patients
Time Frame: 6 months/16 months/28 months after inclusion of last patient
OS rates in MDS-EB2 patients
6 months/16 months/28 months after inclusion of last patient
CR/CRi rates after induction chemotherapy in MDS-EB2 patients
Time Frame: 2 months
CR/CRi rates after induction chemotherapy in MDS-EB2 patients
2 months
CR rates after induction chemotherapy in MDS-EB2 patients
Time Frame: 2 months
CR rates after induction chemotherapy in MDS-EB2 patients
2 months
RFS rates in MDS-EB2 patients
Time Frame: 16 months after inclusion of last patient
RFS rates in MDS-EB2 patients
16 months after inclusion of last patient
CIR rates in MDS-EB2 patients
Time Frame: 16 months after inclusion of last patient
CIR rates in MDS-EB2 patients
16 months after inclusion of last patient
CID rates in MDS-EB2 patients
Time Frame: 16 months after inclusion of last patient
CID rates in MDS-EB2 patients
16 months after inclusion of last patient
EQ-5D-5L questionnaire of the EuroQoL group, among MDS-EB2 patients
Time Frame: 6 months
Health status is measured in terms of five dimensions (5D); mobility, self-care, usual activities, pain/discomfort, and anxiety/depression. Each dimension is rated using a 5 level (5L) scale from 1 (minimum) to 5 (maximum). Higher score values mean a worse outcome.
6 months
European Organisation for Research and Treatment of Cancer Quality of Life Core Questionnaire (EORTC-QLQ-C30) among MDS-EB2 patients
Time Frame: 6 months
All sub scales and single items have values from 1 (minimum) to 100 (maximum) points. A higher point value represents a better condition in terms of the functional scales and quality of life. In the symptomal sub scales a higher point value represents a worse condition.
6 months
Patient Reported Outcome Measurement Information System (PROMIS) Cancer Fatigue short form among MDS-EB2 patients
Time Frame: 6 months
The 7-item PROMIS Cancer Fatigue Short Form assesses the frequency of fatigue in the past seven days (7). Table 2 presents a list of the items. Items are measured on a five point scale (1 = never; 5 = always) and summed, after reverse scoring item 7, with higher scores indicating greater fatigue (worse condition).
6 months
EFS with modified definition
Time Frame: 6 months/16 months after inclusion of last patient

To evaluate the impact of venetoclax on EFS in newly diagnosed AML patients and MDS-EB2 patients using modified endpoint definitions:

o Modified EFS includes treatment failure defined as not achieving CR after induction chemotherapy assessed by investigator. The date of treatment failure is Day 1 post-randomization. Patients achieving CRi are counted as events.

6 months/16 months after inclusion of last patient
RFS with modified definition
Time Frame: 16 months after inclusion of last patient

To evaluate the impact of venetoclax on RFS in newly diagnosed AML patients and MDS-EB2 patients using modified endpoint definitions:

o Modified RFS includes only subjects achieving CR

16 months after inclusion of last patient

Collaborators and Investigators

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

Investigators

  • Principal Investigator: Hartmut Doehner, MD, University of Ulm

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)

September 13, 2022

Primary Completion (Estimated)

February 1, 2025

Study Completion (Estimated)

February 1, 2031

Study Registration Dates

First Submitted

November 3, 2020

First Submitted That Met QC Criteria

November 12, 2020

First Posted (Actual)

November 13, 2020

Study Record Updates

Last Update Posted (Actual)

June 7, 2023

Last Update Submitted That Met QC Criteria

June 6, 2023

Last Verified

June 1, 2023

More Information

Terms related to this study

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 Acute Myeloid Leukemia

Clinical Trials on Venetoclax

3
Subscribe