A Study of ASP2215 in Combination With Induction and Consolidation Chemotherapy in Patients With Newly Diagnosed Acute Myeloid Leukemia.

March 26, 2024 updated by: Astellas Pharma Inc

A Phase 1/2 Study of ASP2215 in Combination With Induction and Consolidation Chemotherapy in Patients With Newly Diagnosed Acute Myeloid Leukemia

The purpose of phase 1 part in this study is to determine the maximum tolerated dose (MTD) and/or recommended expansion dose (RED) of ASP2215 concomitant with cytarabine/idarubicin as induction chemotherapy based on the status of the onset of dose-limiting toxicity (DLT) in newly diagnosed Acute Myeloid Leukemia (AML) subjects. Phase 1 part will also evaluate safety and tolerability and characterize the pharmacokinetic (PK) parameters of ASP2215 concomitant with induction and consolidation chemotherapy as well as evaluate the PK parameters of cytarabine concomitant with ASP2215.

The purpose of phase 2 part is to evaluate efficacy of ASP2215 in combination with induction therapy. Phase 2 cohort will also evaluate safety and characterize the PK parameters of ASP2215 in combination with induction and consolidation therapy followed by maintenance therapy in newly diagnosed FLT3-mutated AML subjects.

Study Overview

Detailed Description

This study is composed of Phase 1 part (the dose-evaluation part and the expansion part) and Phase 2 part.

In the dose-evaluation part of Phase 1 part, at least 3 subjects will receive ASP2215 at each dose (low, middle, and high) for determination of MTD and/or RED. Treatment of AML in Phase 1 part is composed of 3 periods of therapy: remission induction, consolidation, and maintenance. The decision of whether or not to proceed to the next dose will be made based on the occurrence of DLT during Cycle 1 of the induction period.

In the expansion part of Phase 1 part, a maximum of 3 subjects will receive ASP2215 at RED that has been recommended in the dose-evaluation part and the safety will be assessed based on the onset of DLTs during Cycle 1 of the induction and consolidation periods.

In Phase 2 part, Subjects will receive ASP2215 at the recommended dose established in Phase 1 part. The target population will be limited to newly diagnosed FLT3-mutated AML.

Study Type

Interventional

Enrollment (Actual)

84

Phase

  • Phase 2
  • Phase 1

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

      • Akita, Japan
        • Site JP81029
      • Chiba, Japan
        • Site JP81008
      • Fukuoka, Japan
        • Site JP00001
      • Fukuoka, Japan
        • Site JP81004
      • Fukuoka, Japan
        • Site JP81025
      • Fukushima, Japan
        • Site JP81031
      • Gifu, Japan
        • Site JP81030
      • Kochi, Japan
        • Site JP81033
      • Kumamoto, Japan
        • Site JP81028
      • Kyoto, Japan
        • Site JP81016
      • Nagasaki, Japan
        • Site JP81012
      • Okayama, Japan
        • Site JP81009
      • Osaka, Japan
        • Site JP81019
      • Osaka, Japan
        • Site JP81021
    • Aichi
      • Anjo, Aichi, Japan
        • Site JP81037
      • Nagoya, Aichi, Japan
        • Site JP00003
      • Nagoya, Aichi, Japan
        • Site JP81003
      • Nagoya, Aichi, Japan
        • Site JP81027
      • Toyohashi, Aichi, Japan
        • Site JP81038
    • Chiba
      • Narita, Chiba, Japan
        • Site JP81010
    • Ehime
      • Matsuyama, Ehime, Japan
        • Site JP81039
    • Fukui
      • Yoshida-gun, Fukui, Japan
        • Site JP81007
    • Gunma
      • Maebashi, Gunma, Japan
        • Site JP00002
      • Maebshi, Gunma, Japan
        • Site JP81001
    • Hiroshima
      • Fukuyama, Hiroshima, Japan
        • Site JP81026
      • Otake, Hiroshima, Japan
        • Site JP81018
    • Hokkaido
      • Sapporo, Hokkaido, Japan
        • Site JP81014
      • Sapporo, Hokkaido, Japan
        • Site JP81015
    • Hyogo
      • Himeji, Hyogo, Japan
        • Site JP81043
      • Kobe, Hyogo, Japan
        • Site JP00007
      • Kobe, Hyogo, Japan
        • Site JP81006
    • Ibaraki
      • Mito, Ibaraki, Japan
        • Site JP81036
      • Tsukuba, Ibaraki, Japan
        • Site JP81023
    • Ishikawa
      • Kanazawa, Ishikawa, Japan
        • Site JP81020
    • Kanagawa
      • Isehara, Kanagawa, Japan
        • Site JP81013
      • Yokohama, Kanagawa, Japan
        • Site JP00006
      • Yokohama, Kanagawa, Japan
        • Site JP81005
      • Yokohama, Kanagawa, Japan
        • Site JP81024
    • Miyagi
      • Sendai, Miyagi, Japan
        • Site JP81035
    • Nagasaki
      • Omura, Nagasaki, Japan
        • Site JP81011
    • Nara
      • Tenri, Nara, Japan
        • Site JP81041
    • Tochigi
      • Shimono, Tochigi, Japan
        • Site JP81022
    • Tokyo
      • Bunkyo-ku, Tokyo, Japan
        • Site JP81040
      • Shinagawa-ku, Tokyo, Japan
        • Site JP00005
      • Shinagawa-ku, Tokyo, Japan
        • Site JP81032
      • Incheon, Korea, Republic of
        • Site KR82002
      • Seoul, Korea, Republic of
        • Site KR82001
      • Seoul, Korea, Republic of
        • Site KR82005
      • Seoul, Korea, Republic of
        • Site KR82004
      • Seoul, Korea, Republic of
        • Site KR82006
      • Seoul, Korea, Republic of
        • Site KR82003
      • Kaohsiung, Taiwan
        • Site TW88604
      • Taichung, Taiwan
        • Site TW88602
      • Tainan, Taiwan
        • Site TW88601
      • Taoyuan, Taiwan
        • Site TW88603

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 69 years (Adult, Older Adult)

Accepts Healthy Volunteers

No

Description

Inclusion Criteria:

[Phase 1 part]

  • Subject is defined as having previously untreated de novo AML according to the World Health Organization (WHO) criteria (2008) within 28 days prior to study enrollment.
  • Subject has an Eastern Cooperative Oncology Group (ECOG) performance status of ≤ 2.
  • Subject must meet all of the following criteria in the laboratory test at screening:

    • Serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels of ≤ 2.5 × institutional upper limit of normal (ULN)
    • Total serum bilirubin level of ≤ 1.5 × institutional ULN
    • Serum creatinine level of ≤ 1.5 × institutional ULN or an estimated glomerular filtration rate (eGFR) of > 50 mL/min
  • Subject is suitable for oral administration of ASP2215.
  • Female subject falls under the following:

    • Of non-childbearing potential:
    • ・Post-menopausal (defined as at least 1 year with no menses without a medical reason such as drug administration) at screening, or
    • ・Documented surgically sterile or status post-hysterectomy (at least 1 month prior to screening)
    • Of childbearing potential:
    • ・Has a negative result for the pregnancy test at screening, and
    • ・Agrees to use an appropriate contraception starting at screening and throughout the study period and for 60 days after the final study drug administration
  • Female subject agrees not to breastfeed starting at screening and throughout the study period and for 60 days after the final study drug administration.
  • Female subject agrees not to donate ova starting at screening and throughout the study period and for 60 days after the final study drug administration.
  • Male subject and his female spouse/partner who is of childbearing potential agrees to use an appropriate contraception starting at screening and throughout the study period and for 120 days after the final study drug administration.
  • Male subject agrees not to donate sperm starting at screening and throughout the study period and for 120 days after the final study drug administration.
  • Subject agrees not to participate in another interventional study while on study treatment.
  • Subject can be admitted during the induction period.

[Phase 2 part]

  • Subject has a diagnosis of previously-untreated de novo acute myeloid leukemia (AML) according to World Health Organization (WHO) classification (2017) documented within 28 days prior to enrollment.
  • Subject is positive for FLT3-ITD and/or TKD mutation in bone marrow or whole blood as determined by the central lab. Registration by the local lab result is not acceptable.
  • Subject has an ECOG performance status (PS) 0 to 1. Subject who has an ECOG PS 2 is eligible only if the primary disease related symptoms such as pneumonia and febrile neutropenia are the cause of PS score.
  • Subject is suitable for oral administration of ASP2215.
  • Female subject is not pregnant and at least 1 of the following conditions apply:

    • Not a woman of childbearing potential (WOCBP)
    • WOCBP who agrees to follow the contraceptive guidance from the time of informed consent through at least 180 days after final study treatment administration.
  • Female subject must agree not to breastfeed starting at screening and throughout the study period, and for 60 days after the final study drug administration.
  • Female subject must not donate ova starting at screening and throughout the study period, and for 180 days after the final study drug administration.
  • Male subject and their female partners who are of childbearing potential must be using highly effective contraception per locally accepted standards in addition to a barrier method starting at screening and continue throughout the study period and for 120 days after the final study drug administration.
  • Male subject must not donate sperm starting at screening and throughout the study period and for 120 days after the final study drug administration.
  • Male subject with pregnant partner(s) must agree to remain abstinent or use a condom for the duration of the pregnancy throughout the study period and for 120 days after the final study treatment administration.
  • Subject agrees not to participate in another interventional study while on treatment.
  • Subject must meet the following criteria as indicated on the clinical laboratory tests:

    • Serum creatinine ≤ 1.5 × institutional upper limit of normal (ULN), or if serum creatinine outside normal range, then glomerular filtration rate (GFR) > 50 mL/min/1.73m^2 as calculated with the 4-parameter Modification of Diet in Renal Disease (MDRD) equation.
    • Serum total bilirubin ≤ 2.5 mg/dL (43 μmol/L), except for subjects with Gilbert's syndrome.
    • Serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) < 3 x ULN. If liver abnormality by the primary disease is suspected, subject may be pre-registered to initiate the chemotherapy. Prior to registration, AST/ALT values must meet the criteria to continue the study.
    • Serum magnesium ≥ institutional lower limit of normal (LLN). Subject may pre-register without magnesium value, but subject must meet the criteria prior to the full registration on Day 8.
    • Serum potassium ≥ institutional lower limit of normal (LLN).

Exclusion Criteria:

[Phase 1 part]

  • Subject was diagnosed with acute promyelocytic leukemia (APL).
  • Subject has breakpoint cluster region-abelson (BCR-ABL)-positive leukemia (chronic myelogenous leukemia in blast crisis).
  • Subject has active malignant tumors other than AML or myelodysplastic syndrome (MDS).
  • Subject has received prior AML treatment except for the following:

    • Urgent leukapheresis
    • Hydroxyurea administration for emergency treatment of hyperleukocytosis (≤ 7 days)
    • Administration of retinoic acid before the diagnosis to exclude APL (≤ 7 days)
    • Supportive care using growth factors or cytokines
    • Steroid administration to treat hypersensitivity or blood transfusion reactions
  • Subject has clinically active central nervous system leukemia.
  • Subject has disseminated intravascular coagulation (DIC).
  • Subject has had major surgery within 28 days prior to the first study drug administration.
  • Subject has had radiation therapy within 28 days prior to the first study drug administration.
  • Subject has congestive heart failure of New York Heart Association (NYHA) class 3 or 4, or subject with a past history of congestive heart failure of NYHA class 3 or 4 and in whom echocardiogram (ECHO) or Multiple Gate Acquisition (MUGA) scan performed within 3 months prior to screening or at screening showed a left ventricular ejection fraction (LVEF) of < 45%.
  • Subject has cardiac impairment or a clinically significant cardiac disease, including any one of the following:

    • Complete left bundle branch block
    • Obligate use of a cardiac pacemaker
    • Long QT syndrome at Screening
    • Prolongation of the QTc interval (> 450 ms) on electrocardiogram (ECG) at screening
    • Right bundle branch block + left anterior hemiblock (bifascicular block)
    • Angina pectoris within 3 months prior to study drug administration
    • Acute myocardial infarction within 3 months prior to study drug administration
  • Subject requires treatment with concomitant drugs that are strong inducers of cytochrome P450 (CYP)3A.
  • Subject requires treatment with concomitant drugs that are strong inhibitors or inducers of P glycoprotein (P-gp) with the exception of drugs that are considered absolutely essential for the care of the subject.
  • Subject requires treatment with concomitant drugs that target serotonin 5HT1 or 5HT2B receptors or sigma receptors, with the exception of drugs that are considered absolutely essential for treatment of the subject.
  • Subject has an active uncontrollable infection.
  • Subject is known to have human immunodeficiency virus (HIV) infection.
  • Subject has active hepatitis B or C or other active hepatic disorders.
  • Subject has any condition that, in the investigator's or sub-investigator's opinion, makes the subject unsuitable for study participation.
  • Potassium and magnesium levels of below institutional lower limit of normal in the laboratory test at screening.

[Phase 2 part]

  • Subject was diagnosed with acute promyelocytic leukemia (APL).
  • Subject has known BCR-ABL-positive leukemia (chronic myelogenous leukemia in blast crisis).
  • Subject has therapy-related AML.
  • Subject has active malignant tumors other than AML.
  • Subject has received previous therapy for AML, with the exception of the following:

    • Emergency leukapheresis
    • Emergency treatment for hyperleukocytosis with hydroxyurea for ≤ 10 days
    • Preemptive treatment with retinoic acid prior to exclusion of APL ≤ 7 days
    • Growth factor or cytokine support
    • Steroids for the treatment of hypersensitivity or transfusion reactions.
  • Subject has QTcF interval > 450 ms (average of triplicate determinations based on central reading).
  • Subject with long QT syndrome.
  • Subject has clinically active central nervous system leukemia.
  • Subject has had major surgery within 4 weeks prior to the first study dose.
  • Subject has radiation therapy within 4 weeks prior to the first study dose.
  • Subject has immediate life-threatening, severe complications of leukemia such as severe uncontrolled bleeding and/or severe disseminated intravascular coagulation
  • Subject is known to have human immunodeficiency virus infection.
  • Subject has active hepatitis B or C.
  • Subject has an uncontrolled infection. An infection controlled with an approved or closely monitored antibiotic/antiviral/antifungal treatment is allowed.
  • Subject has uncontrolled angina, severe uncontrolled ventricular arrhythmias, electrocardiographic evidence of acute ischemia, congestive heart failure New York Heart Association (NYHA) class 3 or 4 or subject has a history of congestive heart failure of NYHA class 3 or 4 and echocardiogram (ECHO) or Multiple Gate Acquisition (MUGA) scan performed within 3 months prior to screening or at screening showed a left ventricular ejection fraction (LVEF) of < 45%.
  • Subject requires treatment with concomitant drugs that are strong inducers of cytochrome P450 (CYP) 3A.
  • Subject requires treatment with concomitant drugs that target serotonin 5HT2B receptors or sigma nonspecific receptors, with the exception of drugs that are considered absolutely essential for treatment of the subject.
  • Subject requires treatment with concomitant drugs that are strong inhibitors or inducers of P glycoprotein (P-gp) with the exception of drugs that are considered absolutely essential for the care of the subject.
  • Subject has prior malignancies, except resected basal cell carcinoma or treated cervical carcinoma in situ. Cancer treated with curative intent ≥ 5 years previously will be allowed. Cancer treated with curative intent < 5 years previously will not be allowed.
  • Subject has any condition which makes the subject unsuitable for study participation.

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: Non-Randomized
  • Interventional Model: Sequential Assignment
  • Masking: None (Open Label)

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
Experimental: Phase 1 Dose Evaluation Part
In the dose-evaluation part in Phase 1 part, subjects will receive ASP2215 at assigned single dose for determination of MTD and/or RED. Treatment of AML in this study is composed of 3 periods of therapy: remission induction (42-day cycles x 2 at maximum), consolidation (28-day cycles x 3 at maximum), and maintenance (28-day cycles x 26 at maximum). The decision of whether or not to proceed to the next dose will be made based on the occurrence of DLT during Cycle 1 of the induction period.
Once-daily oral administration on 14 consecutive days in every cycle in each period.
Other Names:
  • ASP2215
  • Xospata
Induction period: Once-daily intravenous injection of 12 mg/m^2 idarubicin on 3 consecutive days.

Induction period: Once-daily intravenous injection of 100 mg/m^2 cytarabine on 7 consecutive days.

Consolidation period: Twice-daily intravenous injection of 1.5 g/m^2 cytarabine on Days 1, 3, and 5.

Experimental: Phase 1 Dose Expansion Part
In the dose expansion part in Phase 1 part, subjects will receive ASP2215 at RED that has been determined in the dose-evaluation part, and the safety will be assessed based on the onset of DLTs during Cycle 1 of the induction and consolidation periods.
Once-daily oral administration on 14 consecutive days in every cycle in each period.
Other Names:
  • ASP2215
  • Xospata
Induction period: Once-daily intravenous injection of 12 mg/m^2 idarubicin on 3 consecutive days.

Induction period: Once-daily intravenous injection of 100 mg/m^2 cytarabine on 7 consecutive days.

Consolidation period: Twice-daily intravenous injection of 1.5 g/m^2 cytarabine on Days 1, 3, and 5.

Experimental: Phase 2 Part
Subjects will receive ASP2215 at the recommended dose established in Phase 1 part.
Once-daily oral administration on 14 consecutive days in every cycle in each period.
Other Names:
  • ASP2215
  • Xospata
Induction period: Once-daily intravenous injection of 12 mg/m^2 idarubicin on 3 consecutive days.

Induction period: Once-daily intravenous injection of 100 mg/m^2 cytarabine on 7 consecutive days.

Consolidation period: Twice-daily intravenous injection of 1.5 g/m^2 cytarabine on Days 1, 3, and 5.

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Phase 1 part: Maximum tolerated dose (MTD)
Time Frame: Up to 42 days
MTD is defined as the highest dose of ASP2215 at which the posterior mean of the Dose-Limiting Toxicity (DLT) incidence during Cycle 1 of induction therapy is estimated to be closest to 33%.
Up to 42 days
Phase 1 part: Recommended expansion dose (RED)
Time Frame: Up to 42 days
The sponsor will decide the RED considering the MTD, safety, pharmacokinetics, and efficacy of ASP2215. The final RED will be decided by the sponsor's responsible person by comprehensively assessing the data obtained from the study, and taking into account the discussion held between the sponsor, the medical expert, the investigator, and the advisor of medical statistics.
Up to 42 days
Phase 1 part: Number of participants with dose limiting toxicities (DLTs)
Time Frame: Up to 42 days
A DLT is defined as any Grade ≥ 3 non-hematologic or extramedullary toxicity or any events that require dose reduction of ASP2215 that occur during the DLT assessment period, and that is considered to be possibly, probably, or definitely related to induction or consolidation therapies including the study drugs. The DLT assessment period for making a decision of whether or not to proceed to the next dose is defined as the shorter of the following 2 periods: 39 days from the start of the treatment with ASP2215 in the induction period or days between the start of induction therapy and the start of the first consolidation therapy. For safety assessment during the expansion part, the DLT assessment period includes Cycle 1 of consolidation therapy in addition to the period defined above.
Up to 42 days
Phase 1 part: Number of participants with Adverse Events (AEs)
Time Frame: Up to 9 months

AEs will be coded using Medical Dictionary for Regulatory Activities (MedDRA). A treatment-emergent adverse event (TEAE) is defined as an AE observed after starting administration of the investigational product (IP) and within 28 days after the last administration of IP for phase 1 part. An IP-related TEAE is defined as any TEAE with a causal relationship assessed as YES by the investigator.

AE is considered "serious" if the investigator or sponsor view any of the following outcomes: Death, life-threatening, persistent or significant disability/incapacity, congenital anomaly or birth defect, hospitalization, or medically important event.

Up to 9 months
Phase 1 part: Number of participants with laboratory value abnormalities and/or AEs
Time Frame: Up to 9 months
Number of participants with potentially clinically significant laboratory values.
Up to 9 months
Phase 1 part: Number of participants with vital sign abnormalities and/or AEs
Time Frame: Up to 9 months
Number of participants with potentially clinically significant vital sign values.
Up to 9 months
Phase 2 part: Complete remission (CR) rate after induction therapy period
Time Frame: Up to 4 months
CR is defined as a morphologically leukemia-free state at the post-baseline visit, having a neutrophil count of ≥ 1,000/mm^3 and platelet count of ≥ 100,000/mm^3, bone marrow blasts < 5%. There must be no evidence of Auer rods and no evidence of extramedullary leukemia. The blast counts in peripheral blood must be ≤ 2%.
Up to 4 months

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Phase 1 part: Pharmacokinetics of ASP2215 in plasma: Maximum concentration (Cmax)
Time Frame: Up to 9 months
Cmax will be recorded from the PK plasma samples collected.
Up to 9 months
Phase 1 part: PK of ASP2215 in plasma: Time to attain Cmax (tmax)
Time Frame: Up to 9 months
tmax will be recorded from the PK plasma samples collected.
Up to 9 months
Phase 1 part: PK of ASP2215 in plasma: Area under plasma concentration-time curve from time 0 to 24 (AUC24)
Time Frame: Up to 9 months
AUC24 will be recorded from the PK plasma samples collected.
Up to 9 months
Phase 1 part: PK of ASP2215 in plasma: Oral clearance (CL/F)
Time Frame: Up to 9 months
CL/F will be recorded from the PK plasma samples collected.
Up to 9 months
Phase 1 part: PK of ASP2215 in plasma: AUC from time 0 to last measurable concentration (AUClast)
Time Frame: Up to 9 months
AUClast will be recorded from the PK plasma samples collected.
Up to 9 months
Phase 1 part: PK of ASP2215 in plasma: Apparent terminal elimination half-life (t1/2)
Time Frame: Up to 9 months
t1/2 will be recorded from the PK plasma samples collected.
Up to 9 months
Phase 1 part: PK of ASP2215 in plasma: Apparent Volume of Distribution During the Terminal Elimination Phase after Oral Dosing (Vz/F)
Time Frame: Up to 9 months
Vz/F will be recorded from the PK plasma samples collected.
Up to 9 months
Phase 1 part: PK of ASP2215 in plasma: Plasma trough concentration (Ctrough)
Time Frame: Up to 9 months
Ctrough will be recorded from the PK plasma samples collected.
Up to 9 months
Phase 1 part: PK of cytarabine in plasma: Ctrough
Time Frame: Up to 9 months
Ctrough will be recorded from the PK plasma samples collected.
Up to 9 months
Phase 2 part: PK of ASP2215 in plasma: Concentration
Time Frame: Up to 135 days
Concentration will be recorded from the PK plasma samples collected.
Up to 135 days
Phase 2 part: Duration of overall survival (OS)
Time Frame: Up to 41 months
OS is defined as the time from the date of first dose of day 1 to the date of death due to any cause. Patients still alive or lost to follow up will be censored at the time they were last known to be alive.
Up to 41 months
Phase 2 part: Duration of event free survival (EFS)
Time Frame: Up to 41 months
EFS is defined as the time from the date of first dose of study regimen (day 1) until the date of documented relapse, treatment failure or death from any cause, whichever occurs first. For a subject with none of these events, EFS is censored at the date of last disease assessment.
Up to 41 months
Phase 2 part: Duration of relapse free survival (RFS)
Time Frame: Up to 41 months
RFS is defined as time from the date of achievement of first CRc until relapse or death from any cause, whichever comes first. For a subject who is not known to have relapsed or died, RFS is censored on the date of last relapse-free disease assessment date.
Up to 41 months
Phase 2 part: CR rate after each treatment therapy
Time Frame: Up to 34 months
CR is defined as a morphologically leukemia-free state at the post-baseline visit, having a neutrophil count of ≥ 1,000/mm^3 and platelet count of ≥ 100,000/mm^3, bone marrow blasts < 5%. There must be no evidence of Auer rods and no evidence of extramedullary leukemia. The blast counts in peripheral blood must be ≤ 2%.
Up to 34 months
Phase 2 part: CR rate without minimal residual disease (MRD) after each treatment therapy
Time Frame: Up to 34 months
MRD will be measured from bone marrow samples taken at the screening visit, end of treatment/disease progression and from bone marrow samples taken at other time points during the study.
Up to 34 months
Phase 2 part: CR with partial hematological recovery (CRh) rate after each treatment therapy
Time Frame: Up to 34 months
CRh is defined as a condition at the post baseline visit, having bone marrow blasts < 5%, partial hematologic recovery neutrophil count≥ 500/mm^3 and platelet count ≥ 50,000/mm^3, no evidence of extramedullary leukemia and cannot be classified as CR. The blast counts in peripheral blood must be ≤ 2%.
Up to 34 months
Phase 2 part: Composite CR (CRc) rate after each treatment therapy
Time Frame: Up to 34 months

CRc is defined as total of CR, CR with incomplete platelet recovery (CRp) and + CR with incomplete hematologic recovery (CRi).

CRp is defined as a condition that meets all of the CR criteria at the post-baseline visit, except for the unrecovered platelet count (< 100,000/mm^3).

CRi is defined as a condition that meets all of the CR criteria at the post-baseline visit, except for the unrecovered neutrophil count (< 1,000/mm^3; whether or not having unrecovered platelet counts).

Up to 34 months
Phase 2 part: CR/CRh rate after each treatment therapy
Time Frame: Up to 34 months
CR is defined as a morphologically leukemia-free state at the post-baseline visit, having a neutrophil count of ≥ 1,000/mm^3 and platelet count of ≥ 100,000/mm^3, bone marrow blasts < 5%. There must be no evidence of Auer rods and no evidence of extramedullary leukemia. The blast counts in peripheral blood must be ≤ 2%. CRh is defined as a condition at the post baseline visit, having bone marrow blasts < 5%, partial hematologic recovery neutrophil count≥ 500/mm^3 and platelet count ≥ 50,000/mm^3, no evidence of extramedullary leukemia and cannot be classified as CR. The blast counts in peripheral blood must be ≤ 2%.
Up to 34 months
Phase 2 part: Duration of CR
Time Frame: Up to 41 months
Duration of CR is defined as the period from the first day of achieving CR to the first day of confirmed relapse.
Up to 41 months
Phase 2 part: Duration of CR/CRh
Time Frame: Up to 41 months
Duration of CR/CRh is defined as the period from the first day of achieving CR or CRh to the first day of confirmed relapse.
Up to 41 months
Phase 2 part: Duration of CRh
Time Frame: Up to 41 months
Duration of CRh is defined as the period from the first day of achieving CRh to the first day of confirmed relapse.
Up to 41 months
Phase 2 part: Duration of CRc
Time Frame: Up to 41 months
Duration of CRc is defined as the period from the first day of achieving CRc to the first day of confirmed relapse.
Up to 41 months
Phase 2 part: Duration of response
Time Frame: Up to 41 months
Duration of response is defined as the period from the first day of achieving CR, CRp, CRi, or PR to the first day of confirmed relapse.
Up to 41 months
Phase 2 part: Number of participants with AEs
Time Frame: Up to 35 months

AEs will be coded using MedDRA. A TEAE is defined as an AE observed after starting administration of the IP and within 30 days after the last administration of IP for phase 2 part. An IP-related TEAE is defined as any TEAE with a causal relationship assessed as YES by the investigator.

AE is considered "serious" if the investigator or sponsor view any of the following outcomes: Death, life-threatening, persistent or significant disability/incapacity, congenital anomaly or birth defect, hospitalization, or medically important event.

Up to 35 months
Phase 2 part: Number of participants with laboratory value abnormalities and/or AEs
Time Frame: Up to 35 months
Number of participants with potentially clinically significant laboratory values.
Up to 35 months
Phase 2 part: Number of participants with vital sign abnormalities and/or AEs
Time Frame: Up to 35 months
Number of participants with potentially clinically significant vital sign values.
Up to 35 months
Phase 2 part: Number of participants with 12- ECG abnormalities and/or AEs
Time Frame: Up to 35 months
Number of participants with potentially clinically significant 12-ECG values.
Up to 35 months

Collaborators and Investigators

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

Investigators

  • Study Director: Medical Director, Astellas Pharma Inc

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)

February 26, 2015

Primary Completion (Actual)

August 25, 2021

Study Completion (Estimated)

July 31, 2024

Study Registration Dates

First Submitted

December 1, 2014

First Submitted That Met QC Criteria

December 4, 2014

First Posted (Estimated)

December 8, 2014

Study Record Updates

Last Update Posted (Actual)

March 27, 2024

Last Update Submitted That Met QC Criteria

March 26, 2024

Last Verified

March 1, 2024

More Information

Terms related to this study

Plan for Individual participant data (IPD)

Plan to Share Individual Participant Data (IPD)?

YES

IPD Plan Description

Access to anonymized individual participant level data collected during the study, in addition to study-related supporting documentation, is planned for studies conducted with approved product indications and formulations, as well as compounds terminated during development. Studies conducted with product indications or formulations that remain active in development are assessed after study completion to determine if Individual Participant Data can be shared. Conditions and exceptions are described under the Sponsor Specific Details for Astellas on www.clinicalstudydatarequest.com.

IPD Sharing Time Frame

Access to participant level data is offered to researchers after publication of the primary manuscript (if applicable) and is available as long as Astellas has legal authority to provide the data.

IPD Sharing Access Criteria

Researchers must submit a proposal to conduct a scientifically relevant analysis of the study data. The research proposal is reviewed by an Independent Research Panel. If the proposal is approved, access to the study data is provided in a secure data sharing environment after receipt of a signed Data Sharing Agreement.

IPD Sharing Supporting Information Type

  • STUDY_PROTOCOL
  • SAP
  • CSR

Drug and device information, study documents

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

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 gilteritinib

3
Subscribe