search
Back to results

GM-CLAG in Relapsed/Refractory FLT3-mutated AML

Primary Purpose

Acute Myeloid Leukemia

Status
Withdrawn
Phase
Phase 1
Locations
United States
Study Type
Interventional
Intervention
Gilteritinib 40 MG Oral Tablet
Cladribine
Cytarabine
Filgrastim
Mitoxantrone
Sponsored by
Ayman H Qasrawi
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional treatment trial for Acute Myeloid Leukemia focused on measuring FLT3, CLAG-M, AML, Relapsed, Refractory, Gilteritinib

Eligibility Criteria

18 Years - undefined (Adult, Older Adult)All SexesDoes not accept healthy volunteers

Inclusion Criteria:

  1. Confirmed, morphologically documented AML in first or subsequent relapse or primary induction failure. The diagnosis should be documented within 28 days prior to enrollment.

    1. Relapsed AML is defined as the reappearance of leukemic blasts in the bone marrow, peripheral blood, or any extramedullary site after the attainment of a CR/CRi, detected by morphology, flow cytometry, or immunohistochemistry.
    2. Primary induction failure is defined as failure to achieve a CR or CRi after two courses of induction chemotherapy given per physician's choice or institution's guidelines.
    3. Patients with previous allogeneic hematopoietic stem cell transplantation are allowed to participate, as are prior autologous HCT.
  2. Evidence of FLT3-ITD or FLT3-TKD mutations as detected by polymerase chain reaction and/or next-generation sequencing by peripheral blood and/or bone marrow samples obtained at the time of relapse. FLT3 positivity should be documented before enrollment.
  3. Allowable prior therapies include: First-line AML therapy including cytarabine, anthracyclines, gemtuzumab ozogamicin, prior hypoemethylating agents with or without venetoclax and/or FLT3 inhibitors.
  4. Prior clinical trial participation is allowed with a washout period of experimental drug of at least 4 weeks.
  5. Prior treatment with the FLT3 inhibitor gilteritinib is allowed (must be 12 months or greater from time of eligibility screening).
  6. Age ≥18 years.
  7. ECOG performance status of 0 or 1; Karnofsky 70% or higher (APPENDIX A).
  8. Probability of treatment-related mortality (TRM) <13.1% as calculated by performance status, age, platelet count, albumin, secondary vs primary AML, WBC count, creatinine, and percentage of peripheral blood blasts.50

    a. This score corresponds to corresponding to a ≤13.1% probability of 4-week mortality. Although this score was validated for newly diagnosed AML, it was previously used in the relapsed/refractory setting.30,51,52 (see APPENDIX B). An online calculator is available at: https://cstaging.fhcrc-research.org/TRM/).

  9. Adequate organ function as defined below:

    1. Creatinine clearance (CrCl) >60 mL/min as measured by Cockcroft-Gault formula (APPENDIX C).
    2. Ejection fraction (EF) ≥ 50% as documented by an echocardiogram or multiple gated cardiac blood pool imaging (MUGA) if the echocardiogram's result is judged to be unreliable by the performing cardiologist.
    3. Total bilirubin ≤ 1.5 × the ULN, unless due to Gilbert's, hemolysis or leukemic infiltration, alanine aminotransferase (ALT) and aspartate aminotransferase (AST) ≤ 3 × ULN, unless caused by leukemic infiltration.
  10. Female participants must be either:

    a. Of non-child bearing potential i. postmenopausal status prior to eligibility screening (defined as at least 12 consecutive months of amenorrhea), or ii. undergone successful surgical sterilization (hysterectomy, bilateral tubal ligation, or bilateral oophorectomy).

    b. Of childbearing potential. These women must agree to all of the following: i. Negative serum/urine pregnancy test at eligibility screening. ii. Abstain from pregnancy during the study and for 6 months after the final study drug administration.

    iii. If heterosexually active, agree to consistently use highly effective contraception per locally accepted standards in addition to a barrier method starting at eligibility screening and throughout the study period and for 6 months after the final study drug administration.

  11. Female participants must agree not to breastfeed or donate ova throughout the study drug treatment period and for 6 months after the final study drug administration.
  12. Male patients should agree to:

    1. Avoid impregnating a female partner, during the study and for 180 days after the final study drug administration.
    2. Avoid donating sperm during the study and for 180 days after the final study drug administration.
    3. Consistently use highly effective contraception in addition to a barrier method (even if surgically sterilized) along with their partners who are women of childbearing potential, throughout the study drug treatment period and for 6 months after the final study drug administration.
  13. Ability to understand and willingness to sign a written informed consent document.

Exclusion Criteria:

  1. History of prior treatment with gilteritinib within the last 12 months.
  2. Purine analogue (cladribine and fludarabine) treatment as part of their last line of therapy.
  3. History of allergic reactions attributed to compounds of similar chemical or biologic composition to gilteritinib or other agents used in this study (CLAG-M).
  4. Diagnosis of acute promyelocytic leukemia, BCR/ABL1-positive AML or chronic myelogenous leukemia in blast crisis.
  5. Inability to swallow oral medications or gastrointestinal disease limiting absorption of oral agents.
  6. Patients with a history of allo-SCT should not have active or acute chronic GVHD requiring systemic immunosuppression; topical GVHD treatment is allowed.
  7. Participant has an uncontrolled infection. If a bacterial or viral infection is present, the participant must be receiving definitive therapy and has no signs of progressing infection for 72 hours prior to registration. If a fungal infection is present, the participant must be receiving definitive systemic anti-fungal therapy and have no signs of progressing infection for 1 week prior to registration.

    1. Progressing infection is defined as hemodynamic instability attributable to sepsis or new symptoms, worsening physical signs or radiographic findings attributable to infection.
    2. Persisting fever without other signs or symptoms will not be interpreted as progressing infection.
  8. Active hepatitis B virus (HBV) infection as evidenced by positive hepatitis B surface antigen and/or Nucleic Acid Amplification Testing (NAAT). Patients with prior history of cleared HBV infection (negative hepatitis B surface antigen, positive hepatitis B surface antibody, and positive hepatitis B core IgG antibody) could be allowed to participate after consultation with a hepatologist, in which case Hep B viral load will be monitored by qPCR per institutional guidelines. In this case, concomitant suppressive antiviral therapy against HBV might be warranted.
  9. Uncontrolled HIV infection. Patients who have controlled infection on antiretroviral therapy are allowed, defined as HIV RNA below level of detection for the institution's standard assay.
  10. History of heart failure, myocarditis, or cardiomyopathy which requires heart failure directed therapy.
  11. Participant has long QT Syndrome at screening.
  12. Participant has a Fridericia-corrected QT interval (QTcF) > 450 msec (average of triplicate determinations). Prolonged QTc interval associated with bundle branch block or pacemaking is permitted with written approval of the Principal Investigator (PI).
  13. Untreated CNS leukemia. Evaluation of cerebrospinal fluid (CSF) or brain MRI during screening is only required if there is a clinical suspicion of CNS involvement by leukemia during screening. Patients who previously had CNS leukemia that resolved with therapy should undergo CSF sampling +/- brain MRI within 2 weeks prior to enrollment to exclude CNS relapse.
  14. History of auto-immune conditions requiring active immunosuppressive therapy.
  15. Patients with uncontrolled intercurrent illness including, but not limited to, symptomatic decompensated congestive heart failure; unstable angina pectoris; cardiac arrhythmia or any acute pulmonary condition requiring need for supplemental oxygen.
  16. Participant is receiving concomitant drugs that are strong inducers of CYP3A except for drugs considered essential for the care of the patient.
  17. Any medical condition which, in the investigator's opinion, could compromise the patient's safety.
  18. Patients with psychiatric illness/social situations that would limit compliance with study requirements.
  19. Pregnant or lactating women.
  20. Participants who are receiving any other investigational treatment agents.

Sites / Locations

  • University of Kentucky

Arms of the Study

Arm 1

Arm 2

Arm 3

Arm Type

Experimental

Experimental

Experimental

Arm Label

Gilteritinib (Dose Level -1: 40 mg/day)

Gilteritinib (Dose Level 1: 80 mg/day)

Gilteritinib (Dose Level 2: 120 mg/day)

Arm Description

Dose escalation for gilteritinib will be conducted according to a BOIN design. Gilteritinib 40 mg/day will be given orally starting day 6 until day 19. CLAG-M chemotherapy will be administered at a fixed dose and schedule as following: Cladribine (CL) 5 mg/m2 I.V. over 2 hours once per day on days 1 to 5, given first. Cytarabine (A) 2,000 mg/m2 I.V. over 4 hours once per day on days 1 to 5, given second, 2 hours after cladribine. Filgrastim (GCSF) 300 mcg S.C. once per day on days 0 to 5, started 24 hours prior to chemotherapy. If WBC is > 20 × 109/L on day 0, the filgrastim dose on day 0 will be omitted. Mitoxantrone (M) 10 mg/m2 I.V. once per day on days 1 to 3

Dose escalation for gilteritinib will be conducted according to a BOIN design. Gilteritinib 80 mg/day will be given orally starting day 6 until day 19. CLAG-M chemotherapy will be administered at a fixed dose and schedule as following: Cladribine (CL) 5 mg/m2 I.V. over 2 hours once per day on days 1 to 5, given first. Cytarabine (A) 2,000 mg/m2 I.V. over 4 hours once per day on days 1 to 5, given second, 2 hours after cladribine. Filgrastim (GCSF) 300 mcg S.C. once per day on days 0 to 5, started 24 hours prior to chemotherapy. If WBC is > 20 × 109/L on day 0, the filgrastim dose on day 0 will be omitted. Mitoxantrone (M) 10 mg/m2 I.V. once per day on days 1 to 3

Dose escalation for gilteritinib will be conducted according to a BOIN design. Gilteritinib 120 mg/day will be given orally starting day 6 until day 19. CLAG-M chemotherapy will be administered at a fixed dose and schedule as following: Cladribine (CL) 5 mg/m2 I.V. over 2 hours once per day on days 1 to 5, given first. Cytarabine (A) 2,000 mg/m2 I.V. over 4 hours once per day on days 1 to 5, given second, 2 hours after cladribine. Filgrastim (GCSF) 300 mcg S.C. once per day on days 0 to 5, started 24 hours prior to chemotherapy. If WBC is > 20 × 109/L on day 0, the filgrastim dose on day 0 will be omitted. Mitoxantrone (M) 10 mg/m2 I.V. once per day on days 1 to 3

Outcomes

Primary Outcome Measures

Dose-limiting toxicities (DLT) of gilteritinib when combined with mitoxantrone, cladribine, cytarabine and filgrastim (GM-CLAG)
DLT is defined as: Any treatment-related death Grade 4 neutropenia or thrombocytopenia lasting > 42 days from start of cycle in the absence of active AML Any Grade 4 or greater ANC lasting past cycle day 42 Any Grade 4 or greater platelets lasting past cycle day 42 Grade ≥ 4 organ toxicity (i.e., adverse reactions involving neurologic, pulmonary, cardiac, gastrointestinal, genitourinary, renal, hepatic, or cutaneous systems.) Confirmed Hy's law cases
Maximally Tolerated Dose (MTD) of gilteritinib when combined with mitoxantrone, cladribine, cytarabine and filgrastim (GM-CLAG)
The MTD will be defined as the highest dose of Gilteritinib estimated to have less than 20% hematologic or non-hematologic DLT rates. Enrollment of next patient will be allowed only after prior cohort has completed Cycle 1 and recovered or failure of response has been documented by Day 42.

Secondary Outcome Measures

Complete remission (CR) rate after completing induction chemotherapy with the GM-CLAG combination.
All the following criteria should be met: bone marrow blasts <5% absence of circulating blasts and blasts with Auer roda absence of extramedullary disease ANC ≥ 1.0 × 109 /L (1000/µL) platelet count ≥ 100 × 109 /L (100,000/µL).
Complete remission with incomplete hematologic recovery (CRi) after completing induction chemotherapy with the GM-CLAG combination.
All CR criteria should be met except for residual neutropenia ANC ≤ 1.0 × 109 /L (1000/µL) or thrombocytopenia ≤ 100 × 109 /L (100,000 /µL).
Complete remission with partial hematologic recovery (CRh) after completing induction chemotherapy with the GM-CLAG combination.
All CR criteria should be met except for residual neutropenia and thrombocytopenia with at least partial recovery as evidenced by: ANC ≥ 0.5 × 109 /L (500/µL) and platelet count ≥ 50 × 109 /L (100,000/µL).
Composite complete remission (CRc) after completing induction chemotherapy with the GM-CLAG combination.
This is a composite rate of subjects who achieve a CR and those who achieve CRi.
Minimal residual disease status (MRD) after completing induction chemotherapy with the GM-CLAG combination.
All hematologic criteria of CR; decrease of bone marrow blast percentage to 5% to 25%; MRD status will be determined by FLT3 polymerase chain reaction (PCR) testing of bone marrow aspirates obtained at the end of each cycle of induction.
Best response to Induction Therapy
Best response is defined as the best measured response (CR, CRp, CRi, or PR) post-treatment. Two best responses, up to the time of 2 cycles of treatment period will be defined.
Percentage of patients proceeding to transplantation
To evaluate percentage of patients proceeding to transplantation at anytime during study period
Overall survival (OS)
Defined for all patients of the trial; measured from the date of registration to the date of death from any cause; patients not known to have died at last follow-up are censored on the date they were last known to be alive.
Leukemia-free survival (LFS)
Defined only for patients achieving CR, or CRi; measured from the date of achievement of a remission until the date of relapse or death from any cause.

Full Information

First Posted
April 8, 2022
Last Updated
April 21, 2023
Sponsor
Ayman H Qasrawi
Collaborators
Astellas Pharma Inc
search

1. Study Identification

Unique Protocol Identification Number
NCT05330377
Brief Title
GM-CLAG in Relapsed/Refractory FLT3-mutated AML
Official Title
Phase Ib Trial of Gilteritinib in Combination With Mitoxantrone, Cladribine, Cytarabine and Filgrastim (GM-CLAG) for Relapsed/Refractory FLT3-mutated Acute Myeloid Leukemia
Study Type
Interventional

2. Study Status

Record Verification Date
April 2023
Overall Recruitment Status
Withdrawn
Why Stopped
Sponsor withdrew funding
Study Start Date
March 2023 (Anticipated)
Primary Completion Date
May 30, 2027 (Anticipated)
Study Completion Date
May 30, 2032 (Anticipated)

3. Sponsor/Collaborators

Responsible Party, by Official Title
Sponsor-Investigator
Name of the Sponsor
Ayman H Qasrawi
Collaborators
Astellas Pharma Inc

4. Oversight

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
Data Monitoring Committee
Yes

5. Study Description

Brief Summary
The purpose of this study is to evaluate the dose-limiting toxicities (DLT) and define the maximum tolerated dose (MTD) and the recommended phase II study dose of gilteritinib when combined with mitoxantrone, cladribine, cytarabine and filgrastim (GM-CLAG) in participants with FLT3- mutated relapsed or refractory (R/R) acute myeloid leukemia (AML).
Detailed Description
The treatment of FLT3- mutated relapsed or refractory (R/R) is challenging. Gilteritinib, as a single agent, is the first FLT3 inhibitor to be FDA approved in the R/R setting. This phase I clinical trial will evaluate DLT/MTD and the recommended phase II study dose of gilteritinib when combined with mitoxantrone, cladribine, cytarabine and filgrastim (CLAG-M) in patients with FLT3- mutated R/R AML. This study will also evaluate the pharmacodynamics and pharmacokinetics of gilteritinib when combined with CLAG-M at specific time points. Although this combination has not been established to have superior clinical benefit in comparison to single-agent gilteritinib, the objective of this trial is to provide a possible therapeutic benefit in addition to safety and tolerability.

6. Conditions and Keywords

Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
Acute Myeloid Leukemia
Keywords
FLT3, CLAG-M, AML, Relapsed, Refractory, Gilteritinib

7. Study Design

Primary Purpose
Treatment
Study Phase
Phase 1
Interventional Study Model
Single Group Assignment
Masking
None (Open Label)
Allocation
Non-Randomized
Enrollment
0 (Actual)

8. Arms, Groups, and Interventions

Arm Title
Gilteritinib (Dose Level -1: 40 mg/day)
Arm Type
Experimental
Arm Description
Dose escalation for gilteritinib will be conducted according to a BOIN design. Gilteritinib 40 mg/day will be given orally starting day 6 until day 19. CLAG-M chemotherapy will be administered at a fixed dose and schedule as following: Cladribine (CL) 5 mg/m2 I.V. over 2 hours once per day on days 1 to 5, given first. Cytarabine (A) 2,000 mg/m2 I.V. over 4 hours once per day on days 1 to 5, given second, 2 hours after cladribine. Filgrastim (GCSF) 300 mcg S.C. once per day on days 0 to 5, started 24 hours prior to chemotherapy. If WBC is > 20 × 109/L on day 0, the filgrastim dose on day 0 will be omitted. Mitoxantrone (M) 10 mg/m2 I.V. once per day on days 1 to 3
Arm Title
Gilteritinib (Dose Level 1: 80 mg/day)
Arm Type
Experimental
Arm Description
Dose escalation for gilteritinib will be conducted according to a BOIN design. Gilteritinib 80 mg/day will be given orally starting day 6 until day 19. CLAG-M chemotherapy will be administered at a fixed dose and schedule as following: Cladribine (CL) 5 mg/m2 I.V. over 2 hours once per day on days 1 to 5, given first. Cytarabine (A) 2,000 mg/m2 I.V. over 4 hours once per day on days 1 to 5, given second, 2 hours after cladribine. Filgrastim (GCSF) 300 mcg S.C. once per day on days 0 to 5, started 24 hours prior to chemotherapy. If WBC is > 20 × 109/L on day 0, the filgrastim dose on day 0 will be omitted. Mitoxantrone (M) 10 mg/m2 I.V. once per day on days 1 to 3
Arm Title
Gilteritinib (Dose Level 2: 120 mg/day)
Arm Type
Experimental
Arm Description
Dose escalation for gilteritinib will be conducted according to a BOIN design. Gilteritinib 120 mg/day will be given orally starting day 6 until day 19. CLAG-M chemotherapy will be administered at a fixed dose and schedule as following: Cladribine (CL) 5 mg/m2 I.V. over 2 hours once per day on days 1 to 5, given first. Cytarabine (A) 2,000 mg/m2 I.V. over 4 hours once per day on days 1 to 5, given second, 2 hours after cladribine. Filgrastim (GCSF) 300 mcg S.C. once per day on days 0 to 5, started 24 hours prior to chemotherapy. If WBC is > 20 × 109/L on day 0, the filgrastim dose on day 0 will be omitted. Mitoxantrone (M) 10 mg/m2 I.V. once per day on days 1 to 3
Intervention Type
Drug
Intervention Name(s)
Gilteritinib 40 MG Oral Tablet
Other Intervention Name(s)
Xospata
Intervention Description
Gilteritinib is an oral FLT3 inhibitor, and is active against both FLT3-ITD and FLT3-TKD mutations. Gilteritinib will be given orally according to the assigned phase I dose cohort starting day 6 until day 19. Participants will be given the daily dose with water as at the same time each morning. Gilteritinib should be taken at least 2 hours after or 1 hour before consuming food.
Intervention Type
Drug
Intervention Name(s)
Cladribine
Other Intervention Name(s)
Leustatin
Intervention Description
Cladribine is given intravenously at a dose of 5 mg/m2 I.V. over 2 hours once per day on days 1 to 5.
Intervention Type
Drug
Intervention Name(s)
Cytarabine
Other Intervention Name(s)
cytosine arabinoside (Ara-C)
Intervention Description
Cytarabine is given intravenously at a dose of 2,000 mg/m2 I.V. over 4 hours once per day on days 1 to 5, given second, 2 hours after cladribine.
Intervention Type
Drug
Intervention Name(s)
Filgrastim
Other Intervention Name(s)
GCSF, Filgrastim sndz
Intervention Description
Filgrastim (GCSF) or biosimilar, will be given at a dose of 300 mcg S.C. once per day on days 0 to 5, started 24 hours prior to chemotherapy. If WBC is > 20 × 109/L on day 0, the filgrastim dose on day 0 will be omitted.
Intervention Type
Drug
Intervention Name(s)
Mitoxantrone
Other Intervention Name(s)
Novantrone
Intervention Description
Mitoxantrone is given intravenously at a dose of 10 mg/m2 I.V. over 2 hours once per day on days 1 to 3.
Primary Outcome Measure Information:
Title
Dose-limiting toxicities (DLT) of gilteritinib when combined with mitoxantrone, cladribine, cytarabine and filgrastim (GM-CLAG)
Description
DLT is defined as: Any treatment-related death Grade 4 neutropenia or thrombocytopenia lasting > 42 days from start of cycle in the absence of active AML Any Grade 4 or greater ANC lasting past cycle day 42 Any Grade 4 or greater platelets lasting past cycle day 42 Grade ≥ 4 organ toxicity (i.e., adverse reactions involving neurologic, pulmonary, cardiac, gastrointestinal, genitourinary, renal, hepatic, or cutaneous systems.) Confirmed Hy's law cases
Time Frame
42 days
Title
Maximally Tolerated Dose (MTD) of gilteritinib when combined with mitoxantrone, cladribine, cytarabine and filgrastim (GM-CLAG)
Description
The MTD will be defined as the highest dose of Gilteritinib estimated to have less than 20% hematologic or non-hematologic DLT rates. Enrollment of next patient will be allowed only after prior cohort has completed Cycle 1 and recovered or failure of response has been documented by Day 42.
Time Frame
42 days
Secondary Outcome Measure Information:
Title
Complete remission (CR) rate after completing induction chemotherapy with the GM-CLAG combination.
Description
All the following criteria should be met: bone marrow blasts <5% absence of circulating blasts and blasts with Auer roda absence of extramedullary disease ANC ≥ 1.0 × 109 /L (1000/µL) platelet count ≥ 100 × 109 /L (100,000/µL).
Time Frame
42 days
Title
Complete remission with incomplete hematologic recovery (CRi) after completing induction chemotherapy with the GM-CLAG combination.
Description
All CR criteria should be met except for residual neutropenia ANC ≤ 1.0 × 109 /L (1000/µL) or thrombocytopenia ≤ 100 × 109 /L (100,000 /µL).
Time Frame
42 days
Title
Complete remission with partial hematologic recovery (CRh) after completing induction chemotherapy with the GM-CLAG combination.
Description
All CR criteria should be met except for residual neutropenia and thrombocytopenia with at least partial recovery as evidenced by: ANC ≥ 0.5 × 109 /L (500/µL) and platelet count ≥ 50 × 109 /L (100,000/µL).
Time Frame
42 days
Title
Composite complete remission (CRc) after completing induction chemotherapy with the GM-CLAG combination.
Description
This is a composite rate of subjects who achieve a CR and those who achieve CRi.
Time Frame
42 days
Title
Minimal residual disease status (MRD) after completing induction chemotherapy with the GM-CLAG combination.
Description
All hematologic criteria of CR; decrease of bone marrow blast percentage to 5% to 25%; MRD status will be determined by FLT3 polymerase chain reaction (PCR) testing of bone marrow aspirates obtained at the end of each cycle of induction.
Time Frame
Two Induction Cycles (each up to 42 days)
Title
Best response to Induction Therapy
Description
Best response is defined as the best measured response (CR, CRp, CRi, or PR) post-treatment. Two best responses, up to the time of 2 cycles of treatment period will be defined.
Time Frame
Two Induction Cycles (each up to 42 days)
Title
Percentage of patients proceeding to transplantation
Description
To evaluate percentage of patients proceeding to transplantation at anytime during study period
Time Frame
Up to 2 years
Title
Overall survival (OS)
Description
Defined for all patients of the trial; measured from the date of registration to the date of death from any cause; patients not known to have died at last follow-up are censored on the date they were last known to be alive.
Time Frame
Up to 2 years
Title
Leukemia-free survival (LFS)
Description
Defined only for patients achieving CR, or CRi; measured from the date of achievement of a remission until the date of relapse or death from any cause.
Time Frame
Up to 2 years

10. Eligibility

Sex
All
Minimum Age & Unit of Time
18 Years
Accepts Healthy Volunteers
No
Eligibility Criteria
Inclusion Criteria: Confirmed, morphologically documented AML in first or subsequent relapse or primary induction failure. The diagnosis should be documented within 28 days prior to enrollment. Relapsed AML is defined as the reappearance of leukemic blasts in the bone marrow, peripheral blood, or any extramedullary site after the attainment of a CR/CRi, detected by morphology, flow cytometry, or immunohistochemistry. Primary induction failure is defined as failure to achieve a CR or CRi after two courses of induction chemotherapy given per physician's choice or institution's guidelines. Patients with previous allogeneic hematopoietic stem cell transplantation are allowed to participate, as are prior autologous HCT. Evidence of FLT3-ITD or FLT3-TKD mutations as detected by polymerase chain reaction and/or next-generation sequencing by peripheral blood and/or bone marrow samples obtained at the time of relapse. FLT3 positivity should be documented before enrollment. Allowable prior therapies include: First-line AML therapy including cytarabine, anthracyclines, gemtuzumab ozogamicin, prior hypoemethylating agents with or without venetoclax and/or FLT3 inhibitors. Prior clinical trial participation is allowed with a washout period of experimental drug of at least 4 weeks. Prior treatment with the FLT3 inhibitor gilteritinib is allowed (must be 12 months or greater from time of eligibility screening). Age ≥18 years. ECOG performance status of 0 or 1; Karnofsky 70% or higher (APPENDIX A). Probability of treatment-related mortality (TRM) <13.1% as calculated by performance status, age, platelet count, albumin, secondary vs primary AML, WBC count, creatinine, and percentage of peripheral blood blasts.50 a. This score corresponds to corresponding to a ≤13.1% probability of 4-week mortality. Although this score was validated for newly diagnosed AML, it was previously used in the relapsed/refractory setting.30,51,52 (see APPENDIX B). An online calculator is available at: https://cstaging.fhcrc-research.org/TRM/). Adequate organ function as defined below: Creatinine clearance (CrCl) >60 mL/min as measured by Cockcroft-Gault formula (APPENDIX C). Ejection fraction (EF) ≥ 50% as documented by an echocardiogram or multiple gated cardiac blood pool imaging (MUGA) if the echocardiogram's result is judged to be unreliable by the performing cardiologist. Total bilirubin ≤ 1.5 × the ULN, unless due to Gilbert's, hemolysis or leukemic infiltration, alanine aminotransferase (ALT) and aspartate aminotransferase (AST) ≤ 3 × ULN, unless caused by leukemic infiltration. Female participants must be either: a. Of non-child bearing potential i. postmenopausal status prior to eligibility screening (defined as at least 12 consecutive months of amenorrhea), or ii. undergone successful surgical sterilization (hysterectomy, bilateral tubal ligation, or bilateral oophorectomy). b. Of childbearing potential. These women must agree to all of the following: i. Negative serum/urine pregnancy test at eligibility screening. ii. Abstain from pregnancy during the study and for 6 months after the final study drug administration. iii. If heterosexually active, agree to consistently use highly effective contraception per locally accepted standards in addition to a barrier method starting at eligibility screening and throughout the study period and for 6 months after the final study drug administration. Female participants must agree not to breastfeed or donate ova throughout the study drug treatment period and for 6 months after the final study drug administration. Male patients should agree to: Avoid impregnating a female partner, during the study and for 180 days after the final study drug administration. Avoid donating sperm during the study and for 180 days after the final study drug administration. Consistently use highly effective contraception in addition to a barrier method (even if surgically sterilized) along with their partners who are women of childbearing potential, throughout the study drug treatment period and for 6 months after the final study drug administration. Ability to understand and willingness to sign a written informed consent document. Exclusion Criteria: History of prior treatment with gilteritinib within the last 12 months. Purine analogue (cladribine and fludarabine) treatment as part of their last line of therapy. History of allergic reactions attributed to compounds of similar chemical or biologic composition to gilteritinib or other agents used in this study (CLAG-M). Diagnosis of acute promyelocytic leukemia, BCR/ABL1-positive AML or chronic myelogenous leukemia in blast crisis. Inability to swallow oral medications or gastrointestinal disease limiting absorption of oral agents. Patients with a history of allo-SCT should not have active or acute chronic GVHD requiring systemic immunosuppression; topical GVHD treatment is allowed. Participant has an uncontrolled infection. If a bacterial or viral infection is present, the participant must be receiving definitive therapy and has no signs of progressing infection for 72 hours prior to registration. If a fungal infection is present, the participant must be receiving definitive systemic anti-fungal therapy and have no signs of progressing infection for 1 week prior to registration. Progressing infection is defined as hemodynamic instability attributable to sepsis or new symptoms, worsening physical signs or radiographic findings attributable to infection. Persisting fever without other signs or symptoms will not be interpreted as progressing infection. Active hepatitis B virus (HBV) infection as evidenced by positive hepatitis B surface antigen and/or Nucleic Acid Amplification Testing (NAAT). Patients with prior history of cleared HBV infection (negative hepatitis B surface antigen, positive hepatitis B surface antibody, and positive hepatitis B core IgG antibody) could be allowed to participate after consultation with a hepatologist, in which case Hep B viral load will be monitored by qPCR per institutional guidelines. In this case, concomitant suppressive antiviral therapy against HBV might be warranted. Uncontrolled HIV infection. Patients who have controlled infection on antiretroviral therapy are allowed, defined as HIV RNA below level of detection for the institution's standard assay. History of heart failure, myocarditis, or cardiomyopathy which requires heart failure directed therapy. Participant has long QT Syndrome at screening. Participant has a Fridericia-corrected QT interval (QTcF) > 450 msec (average of triplicate determinations). Prolonged QTc interval associated with bundle branch block or pacemaking is permitted with written approval of the Principal Investigator (PI). Untreated CNS leukemia. Evaluation of cerebrospinal fluid (CSF) or brain MRI during screening is only required if there is a clinical suspicion of CNS involvement by leukemia during screening. Patients who previously had CNS leukemia that resolved with therapy should undergo CSF sampling +/- brain MRI within 2 weeks prior to enrollment to exclude CNS relapse. History of auto-immune conditions requiring active immunosuppressive therapy. Patients with uncontrolled intercurrent illness including, but not limited to, symptomatic decompensated congestive heart failure; unstable angina pectoris; cardiac arrhythmia or any acute pulmonary condition requiring need for supplemental oxygen. Participant is receiving concomitant drugs that are strong inducers of CYP3A except for drugs considered essential for the care of the patient. Any medical condition which, in the investigator's opinion, could compromise the patient's safety. Patients with psychiatric illness/social situations that would limit compliance with study requirements. Pregnant or lactating women. Participants who are receiving any other investigational treatment agents.
Overall Study Officials:
First Name & Middle Initial & Last Name & Degree
Ayman Qasrawi, MD
Organizational Affiliation
University of Kentucky
Official's Role
Principal Investigator
Facility Information:
Facility Name
University of Kentucky
City
Lexington
State/Province
Kentucky
ZIP/Postal Code
40536
Country
United States

12. IPD Sharing Statement

Plan to Share IPD
No

Learn more about this trial

GM-CLAG in Relapsed/Refractory FLT3-mutated AML

We'll reach out to this number within 24 hrs