Fludarabine in Combination With Daunorubicin and Cytarabine Liposome in Newly-diagnosed Acute Myeloid Leukemia.

September 19, 2022 updated by: James Mangan, University of California, San Diego

A Phase II Trial of Fludarabine in Combination With Daunorubicin and Cytarabine Liposome for Adults With Newly-diagnosed Acute Myeloid Leukemia: University of California Hematologic Malignancies Consortium Protocol 1914

This phase 2 clinical trial will evaluate the effectiveness and safety of fludarabine in combination with CPX-351 in patients with untreated AML. Patients will receive fludarabine and CPX-351 during Induction 1 and 2 as well as 2 cycles of consolidation therapy.

Study Overview

Status

Terminated

Intervention / Treatment

Detailed Description

This is a phase 2, open label single arm study to look at the effectives and safety of fludarabine in combination with CPX-351 in patients with untreated AML. The rationale for this combination stems from data which indicated that pre-treatment of the THP-1 cell line with fludarabine for 4 hours prior to CPX-351 administration (Flu-CPX) significantly potentiated intracellular ara-CTP accumulation compared to CPX-351 alone. This suggests that fludarabine combined with CPX-351 may have efficacy against leukemic clones that would be resistant to CPX-351 or standard chemotherapy in first induction. It has been demonstrated that treatment with CPX-351 produces superior clinical outcomes in secondary AML likely due to its novel formulation, which results in sustained exposure of the cytotoxic agents cytarabine and daunorubicin in a synergistic 5:1 ratio within the plasma and bone marrow. Fludarabine can potentially improve upon the outcomes observed with CPX-351 monotherapy and 7+3 by enhancing intracellular ara-CTP accumulation from CPX-351. Patients will received fludarabine and CPX-351 for up to 2 cycles of induction and 2 cycles of consolidation.

Study Type

Interventional

Enrollment (Actual)

2

Phase

  • Phase 2

Contacts and Locations

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

Study Locations

    • California
      • La Jolla, California, United States, 92093
        • UCSD Moores Cancer Center

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

Genders Eligible for Study

All

Description

Inclusion Criteria:

  1. Histologically confirmed de novo or secondary AML as defined by WHO criteria
  2. Intermediate- or poor-risk disease by ELN 2017 criteria
  3. Adults 18 years of age or older
  4. ECOG performance status of 0, 1, or 2
  5. Able to give informed consent and follow study guidelines
  6. Organ function requirements:

    1. Adequate renal function defined as creatinine clearance greater than 60 ml/min
    2. Adequate hepatic function defined by serum bilirubin less than or equal 2 mg/dL. If serum bilirubin greater than 2 mg/dl and direct bilirubin is less than 30 percent of total bilirubin contact study chair for eligibility exception for Gilbert's syndrome.
    3. ALT/AST less than or equal to 3 times the upper limit of normal
    4. LVEF 50 percent by echocardiogram or MUGA
  7. Patients with history of second malignancies in complete remission and without history of metastasis are eligible if there is clinical evidence of disease stability for a period of greater than 6 months off cytotoxic chemotherapy, documented by imaging, tumor marker studies, etc., at screening.
  8. Women of child-bearing potential and men with partners of child-bearing potential must agree to use adequate contraception (hormonal or barrier method of birth control; abstinence) prior to study entry, for the duration of study participation, and for 120 days following completion of therapy. Should a woman become pregnant or suspect she is pregnant while participating in this study, she should inform her treating physician immediately.
  9. Women of child-bearing potential has negative pregnancy test prior to initiating study drug dosing.

Exclusion Criteria:

  1. Current or anticipated use of additional investigational agents.
  2. Any prior treatment for AML with the exception of corticosteroids, hydroxyurea, and/or leukapheresis to prevent or treat early complications prior to starting study therapy. Permitted prior therapy must be stopped 24 hours prior to starting study therapy.
  3. Prior use of hypomethylating agents is permitted for patients with history of antecedent MDS. Last dose of hypomethylating therapy must have been 15 or more days prior to starting study therapy. Toxicities associated with prior MDS therapy must have recovered to grade 1 or less prior to start of treatment.
  4. Favorable risk cytogenetics as defined by 2017 ELN risk stratification including acute promyelocytic leukemia
  5. Chronic myeloid leukemia in myeloid blast crisis
  6. Except for CMML, patients with history of myeloproliferative neoplasms (MPN) (defined as a history of essential thrombocytosis or polycythemia vera, or idiopathic myelofibrosis prior to the diagnosis of AML) or combined MDS/MPN are not eligible
  7. Clinical evidence of active CNS leukemia
  8. Active or metastatic second malignancy
  9. Any major surgery or radiation therapy within four weeks.
  10. Patients with prior cumulative anthracycline exposure of greater than 368 mg/m2 daunorubicin (or equivalent).
  11. Any serious medical condition, laboratory abnormality or psychiatric illness that would prevent obtaining informed consent
  12. Patients with myocardial impairment of any cause (e.g. cardiomyopathy, ischemic heart disease, significant valvular dysfunction, hypertensive heart disease, and congestive heart failure) resulting in heart failure by New York Heart Association Criteria (Class III or IV staging)
  13. Active or uncontrolled infection. Patients with an infection receiving treatment (antibiotic, antifungal or antiviral treatment) may be entered into the study but must be afebrile and hemodynamically stable for greater than or equal to 72 hrs.
  14. Current evidence of invasive fungal infection (blood or tissue culture); patients with recent fungal infection must have subsequent negative culture(s) to be eligible
  15. Known HIV infection
  16. Active hepatitis B or hepatitis C infection
  17. Hypersensitivity to cytarabine, daunorubicin or liposomal products
  18. History of Wilson's disease or copper-metabolism disorder
  19. Pregnant or breastfeeding
  20. Any condition which in the opinion of the investigator will interfere with the ability of the subject to comply with the requirements of 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: NA
  • Interventional Model: SINGLE_GROUP
  • Masking: NONE

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
EXPERIMENTAL: Fludarabine and CPX351

Induction 1:

Fludarabine 30 mg/m2/day IV on days 1-5 for 5 doses Daunorubicin and cytarabine liposome (CPX-351) daunorubicin 44 mg/m2/day and cytarabine 100 mg/m2/day IV on days 1, 3, 5 (given 4 hours after fludarabine infusion) for 3 doses

Induction 2 (residual leukemia after Induction 1):

Fludarabine 30 mg/m2/day IV on days 1-3 for 3 doses

Daunorubicin and cytarabine liposome (CPX-351) daunorubicin 44 mg/m2/day and cytarabine 100 mg/m2/day IV on days 1, 3 (given 4 hours after fludarabine infusion) for 2 doses

Optional consolidation, up to 2 cycles:

Daunorubicin and cytarabine liposome (CPX-351) daunorubicin 29 mg/m2/day and cytarabine 65 mg/m2/day IV on days 1, 3 for 2 doses

30mg/m2 days 1 through 5
Other Names:
  • Oforta, Fludara
100U/m2 days 1, 3 5 in induction, 65U/m2 days 1 and 3 for consolidation
Other Names:
  • CPX-351

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Overall response rate after induction
Time Frame: 35 days
Overall response rate after induction, defined as the sum of complete response (CR) rate and complete response with incomplete count recovery (CRi) rate after 1-2 cycles of induction therapy, in accordance with 2017 ELN criteria.
35 days

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Safety and Tolerability
Time Frame: 6 months
Safety and tolerability will be determined by rates of NCI CTCAE 5.0 Grade 3-5 toxicities and rate of discontinuation from study therapy due to intolerance
6 months
Incidence of Grade 3 Treatment Emergent Adverse Events [Safety and Tolerability]
Time Frame: 6 months
Safety and tolerability will be determined by rates of NCI CTCAE 5.0 Grade 3 toxicities and rate of discontinuation from study therapy due to intolerance
6 months
Incidence of Grade 4 Treatment Emergent Adverse Events [Safety and Tolerability]
Time Frame: 6 months
Safety and tolerability will be determined by rates of NCI CTCAE 5.0 Grade 4 toxicities and rate of discontinuation from study therapy due to intolerance
6 months
Incidence of Grade 5 Treatment Emergent Adverse Events [Safety and Tolerability]
Time Frame: 6 months
Safety and tolerability will be determined by rates of NCI CTCAE 5.0 Grade 5 toxicities and rate of discontinuation from study therapy due to intolerance
6 months
CR Rate
Time Frame: 60 days
CR rate defined as proportion of patients achieving a CR after 1-2 cycles of induction
60 days
Overall response rate
Time Frame: 35 days
Overall response rate (CR +CRi) after 1 cycle of induction
35 days
Overall survival
Time Frame: 1 year
Overall survival (OS) at 1 year, with OS defined as time from start of study therapy to death from any cause
1 year
Overall survival
Time Frame: 3 years
Overall survival (OS) at 3 years, with OS defined as time from start of study therapy to death from any cause
3 years
Leukemia-free survival
Time Frame: 1 years
Leukemia-free survival (LFS) at 1 year, with EFS defined as the time from start of study therapy until failure to attain CR, relapse, or death from any cause
1 years
Leukemia-free survival
Time Frame: 3 years
Leukemia-free survival (LFS) at 3 years, with EFS defined as the time from start of study therapy until failure to attain CR, relapse, or death from any cause
3 years
Event free survival
Time Frame: 1 year
Event Free Survival (EFS) at 1 year, with EFS defined as the time from start of study therapy until failure to attain CR, relapse, or death from any cause.
1 year
Event free survival
Time Frame: 3 years
Event Free Survival (EFS) at 3 years, with EFS defined as the time from start of study therapy until failure to attain CR, relapse, or death from any cause.
3 years
Platelet Recovery
Time Frame: 60 days
Platelet recovery, defined as the time from start of study therapy until absolute neutrophil count >1,000/mcl in patients achieving a CR
60 days
30-day
Time Frame: 30 days from start of study therapy
30-day mortality defined as death from any cause within 30 days of starting study therapy
30 days from start of study therapy
60-day mortality
Time Frame: 60 days from start of study therapy
60-day mortality defined as death from any cause within 60 days of starting study therapy
60 days from start of study therapy

Other Outcome Measures

Outcome Measure
Measure Description
Time Frame
Minimal Residual Disease (MRD) Response (positive or negative)
Time Frame: 60 days
MRD response at CR/CRi will be assessed by multiparameter flow cytometry at the University of Washington.
60 days
Descriptive Statistics of Patients Mutation Profile at Screening
Time Frame: At Screening
Somatic mutation profile as determined by next generation sequencing will be performed for recurrent AML mutations using standard technique used at individual sites (local or send-out testing)
At Screening
Descriptive Statistics of Patients Mutation Profile at Relapse
Time Frame: At Relapse
Somatic mutation profile as determined by next generation sequencing will be performed for recurrent AML mutations using standard technique.
At Relapse

Collaborators and Investigators

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

Collaborators

Investigators

  • Study Chair: Matthew Wieduwilt, MD, PhD, UC San Diego

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)

August 5, 2020

Primary Completion (ACTUAL)

July 1, 2022

Study Completion (ACTUAL)

July 1, 2022

Study Registration Dates

First Submitted

June 3, 2020

First Submitted That Met QC Criteria

June 6, 2020

First Posted (ACTUAL)

June 11, 2020

Study Record Updates

Last Update Posted (ACTUAL)

September 22, 2022

Last Update Submitted That Met QC Criteria

September 19, 2022

Last Verified

September 1, 2022

More Information

Terms related to this study

Drug and device information, study documents

Studies a U.S. FDA-regulated drug product

Yes

Studies a U.S. FDA-regulated device product

No

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

Clinical Trials on AML

Clinical Trials on Fludarabine

3
Subscribe