Clinical Trials Logo

Clinical Trial Details — Status: Completed

Administrative data

NCT number NCT02538965
Other study ID # CC-5013-AML-002
Secondary ID
Status Completed
Phase Phase 2
First received
Last updated
Start date November 19, 2015
Est. completion date January 11, 2019

Study information

Verified date December 2019
Source Celgene
Contact n/a
Is FDA regulated No
Health authority
Study type Interventional

Clinical Trial Summary

To determine the activity of lenalidomide in the treatment of pediatric subjects with relapsed/refractory acute myeloid leukemia (AML) (with second or greater relapse or refractory to at least 2 prior induction attempts) measured by morphological complete response defined as either a CR or CRi within the first 4 cycles of treatment.


Description:

This is a multicenter, open-label, single-arm, Phase 2, Simon's Optimal two-stage design study, with an Optional Extension Phase (OEP), that will assess the activity, safety and pharmacokinetics (PK) of lenalidomide in pediatric subjects from 1 to ≤ 18 years of age with second or greater Relapsed or Refractory Acute Myeloid Leukemia (rrAML). A total of 43 evaluable participants (18 participants in Stage 1 and an additional 25 participants in Stage 2) are required for assessment of the primary endpoint. To allow for participants found to be unevaluable for the primary endpoint due to an incorrect diagnosis, not having a disease assessment post screening, or who discontinued prior to receiving lenalidomide, up to 4 additional participants may be enrolled for a maximum of 47 evaluable subjects across approximately 70 sites. Approximately 50% of enrolled participants will be younger than 12 years of age to provide adequate PK data for this age subset.

If during Stage 1, at least 3 of 18 participants achieve a morphologic complete response (either CR or CRi) within the first 4 cycles of study treatment, then the study will proceed to Stage 2; otherwise, the study will be terminated. Similarly, if at the final analysis, at least 8 of 43 evaluable subjects across Stages 1 and 2 achieve a response (CR/CRi) within the first 4 cycles of study treatment, it will be concluded that lenalidomide has sufficient activity in pediatric Acute Myeloid Leukemia (AML) to warrant subsequent study. The optional extension phase (OEP) will allow participants who demonstrate clinical benefit, as assessed by the Investigator at the completion of 12 cycles of lenalidomide therapy, to continue receiving oral lenalidomide until they meet the criteria for study discontinuation. In the OEP, only safety, dosing, concomitant medications/procedures, and second primary malignancies (SPMs) will be monitored.


Recruitment information / eligibility

Status Completed
Enrollment 17
Est. completion date January 11, 2019
Est. primary completion date July 22, 2017
Accepts healthy volunteers No
Gender All
Age group 1 Year to 18 Years
Eligibility Inclusion Criteria:

- Participants must satisfy the following criteria to be enrolled in the study:

1. Male or female is 1 to = 18 years of age at the time of signing the Informed Consent Form / Informed Assent Form (ICF/IAF).

2. Participants (when applicable, parental/legal representative) must understand and voluntarily provide permission to the ICF/IAF prior to conducting any study-related assessments/procedures.

3. Participants have relapsed or refractory acute myeloid leukemia after at least 2 prior induction attempts:

- Bone marrow aspirate or biopsy must have = 5% blasts by morphology and/or flow cytometry.

- Each block of chemotherapy is a separate reinduction attempt.

- Donor lymphocyte infusion (DLI) is considered a reinduction attempt.

4. Participants are willing and able to adhere to the study visit schedule and other protocol requirements.

5. Participants have a Karnofsky score of = 50% (participants = 16 years of age) or a Lansky score = 50% (participants < 16 years of age).

6. Participants have a resting left ventricular ejection fraction (LVEF) of = 40% obtained by echocardiography.

7. Participants have recovered from the acute toxic effects of all prior chemotherapy, immunotherapy, or radiotherapy prior to first dose. All prior treatment-related toxicities must have resolved to = Grade 2 prior to enrollment.

8. Regarding radiation therapy, time elapsed prior to first dose of lenalidomide:

- 2 weeks for local palliative radiation therapy (XRT).

- 8 weeks if prior craniospinal chemoradiation therapy (CRT) or if = 50% radiation of pelvis.

- 6 weeks if other bone marrow radiation has been administered.

9. Graft-versus-host disease criteria:

- Participants must be at least 2 months (from first dose of lenalidomide) from stem cell infusion.

- Participants must have no evidence of active acute or chronic GVHD (Grade 0) for 4 weeks prior to the first dose of lenalidomide.

- If the participants have a history of maximum Grade 1 or 2 GVHD that was treated with systemic steroid (= 0.5 mg/kg/day prednisone equivalents) or other non-steroid systemic IST, the participant must be off all IST for at least 2 weeks, and must have ceased treatment doses of steroids for GVHD (= 0.5 mg/kg/day prednisone equivalents) for at least 4 weeks.

- If the participants have a history of Grade 3 or greater GVHD, the participants must be off all systemic IST for 4 weeks

- Topical therapy is permitted and does not imply the participants have active acute or chronic GVHD.

- Physiologic dosing of hydrocortisone is permitted.

10. At least 4 weeks (from first dose) elapsed from donor lymphocyte infusion (DLI) without conditioning.

11. Participants have adequate renal function, which is defined as:

- Creatinine clearance calculated using the Schwartz formula, or radioisotope glomerular filtration rate (GFR) > 70 mL/min/1.73 m2.

12. Participants have adequate liver function, which is defined as:

- Total bilirubin is = 2 mg/dL unless the increase in bilirubin is attributable to Gilbert's Syndrome

- Aspartate aminotransferase (AST) is = 3.0 x upper normal limit (ULN) for age. For the purpose of this study, the ULN for AST is 50 U/L.

- Alanine transaminase (ALT) is = 3.0 x upper normal limit (ULN) for age. For the purpose of this study, the ULN for ALT is 45 U/L.

13. Female Children of Childbearing Potential (FCCBP), Female of Childbearing Potential (FCBP) and male participants that have reached puberty must agree to undergo physician-approved reproductive education and discuss the side effects of the study therapy on reproduction with parent(s) and/or guardian(s).

14. All participants and/or parents/guardians must have an understanding that lenalidomide could have a potential teratogenic risk. Female children of childbearing potential, is defined as females who have achieved menarche and/or breast development in Tanner Stage 2 or greater and have not undergone a hysterectomy or bilateral oophorectomy and FCBP defined as a sexually mature woman who has not undergone a hysterectomy or bilateral oophorectomy and has not been naturally postmenopausal for at least 24 consecutive months (ie, has had menses at any time in the preceding 24 consecutive months) must meet the following conditions below (Note: Amenorrhea following cancer therapy does not rule out childbearing potential):

- Medically supervised serum pregnancy tests with a sensitivity of at least 25 mIU/mL must be conducted in FCCBP/FCBP, including those who commit to complete abstinence*. FCCBP/FCBP must have two pregnancy tests (with a minimum sensitivity of 25 mIU/mL) prior to starting treatment with lenalidomide. The first pregnancy test must be performed within 10 - 14 days prior to the start of lenalidomide treatment and the second pregnancy test must be performed within 24 hours prior to starting treatment with lenalidomide.

NOTE: The pregnancy test 10 to 14 days prior to initiation of lenalidomide may be omitted, at the discretion of the investigator, for any FCCBP/FCBP who has high acuity disease requiring immediate treatment with lenalidomide. The pregnancy test within 24 hours prior to the first dose of lenalidomide is required to be performed.

The participants may not received Investigational Product (IP) until the investigator has verified that the results of these pregnancy tests performed on Cycle 1 Day 1 are negative. FCCBP/FCBP with regular or no menstrual cycles must agree to have pregnancy tests weekly for the first 28 days of study participation and then every 28 days while on study, at study Treatment Discontinuation Visit, and at Day 28 following IP discontinuation. If menstrual cycles are irregular, the pregnancy testing must occur weekly for the first 28 days and then every 14 days while on study, at study Treatment Discontinuation Visit, and at Days 14 and 28 following IP discontinuation.

- Female participants must, as appropriate to age and at the discretion of the study Investigator, either commit to true abstinence* from heterosexual contact (which must be reviewed on a monthly basis) and/or agree to the use of two reliable forms of approved and effective contraceptive methods simultaneously. The two methods of reliable contraception must include one highly effective method and one additional effective (barrier) method (oral, injectable, or implantable hormonal contraceptive; tubal ligation; intra-uterine device; barrier contraceptive with spermicide; or vasectomized partner) without interruption, 28 days prior to starting lenalidomide treatment, throughout the entire duration of study treatment including dose interruptions and 28 days after the end of study treatment.

- All male and female participants must follow all requirements defined in the Pregnancy Prevention Program.

16. Male participants, as appropriate to age and the discretion of the study physician:

- Must practice true abstinence* or agree to use a condom during sexual contact with a pregnant female or a female of childbearing potential while participating in the study, during dose interruptions and for at least 28 days following lenalidomide discontinuation, even if he has undergone a successful vasectomy or practices complete abstinence.

Exclusion Criteria:

1. Participants have Down syndrome.

2. Participants have French-American-British classification (FAB) type M3 leukemia (acute promyelocytic leukemia) or identification of t(15;17).

3. Participants have isolated central nervous system (CNS) involvement or extramedullary relapse. (Participants with combined CNS/marrow relapse may be enrolled).

4. Participants had prior treatment with cytotoxic chemotherapy within 2 weeks of the first dose of lenalidomide with the exception of hydroxyurea (allowed prior to the first dose of lenalidomide and through Day 14 of Cycle 1) and intrathecal (IT) cytarabine will be administered within 2 weeks prior to administration of lenalidomide.

5. Participants have had prior treatment with biologic antineoplastic agents less than 7 days before the first dose of lenalidomide. For agents that have known adverse events (AEs) occurring beyond 7 days after administration (ie, monoclonal antibodies), this period must be extended beyond the time during which acute AEs are known to occur.

6. Participants have had prior treatment with lenalidomide.

7. Participant is pregnant or lactating.

8. Participants have an uncontrolled systemic fungal, bacterial, or viral infection (defined as ongoing signs/symptoms related to the infection without improvement despite appropriate antibiotics, antiviral therapy, and/or other treatment).

9. Participants has known Human Immunodeficiency Virus (HIV) positivity (participants who are receiving antiretroviral therapy for HIV disease).

10. Participants have a prior history of malignancies other than AML unless the subject has been free of the disease for = 5 years from first dose of lenalidomide.

11. The presence of any of the following will exclude a participant from enrollment:

- Participants have any significant medical condition, laboratory abnormality, or psychiatric illness that would prevent the participant from participating in the study.

- Participants have any condition including the presence of laboratory abnormalities, which places the participant at unacceptable risk if he/she were to participate in the study.

- Participants have any condition that confounds the ability to interpret data from the study.

12. Participants have cardiac disorders (Common Terminology Criteria for Adverse Events [CTCAE] version 4.03 Grade 3 or 4).

13. Participants have a history of well-documented prior veno-occlusive disease (VOD).

14. Participants have any other organ dysfunction (CTCAE version 4.03 Grade 4) that will interfere with the administration of the therapy according to this protocol.

Study Design


Related Conditions & MeSH terms


Intervention

Drug:
Lenalidomide
Lenalidomide will be administered orally once daily for the first 21 days of every 28-day cycle. The starting dose will be 2 mg/kg/day with a maximum dose of 70 mg/day. Number of cycles: 12, or until evidence of progressive disease. Participants will also be discontinued if unresolved toxicities as described in the protocol occur, or if dose reductions are required and subject does not tolerate minimum dose level of 1mg/kg/day.

Locations

Country Name City State
Canada Alberta Childrens Hospital Calgary Alberta
Canada IWK Health Center Halifax Nova Scotia
Canada McGill University Health Center Montreal Quebec
Canada Childrens Hospital of Eastern Ontario Ottawa Ontario
Canada Hospital For Sick Children Torento
Canada British Columbia Children's Hospital Vancouver British Columbia
United States University of Michigan Ann Arbor Michigan
United States Children's Healthcare of Atlanta Atlanta Georgia
United States Colorado Children's Hospital Aurora Colorado
United States Dell Children's Medical Center of Central Texas Austin Texas
United States Johns Hopkins University Baltimore Maryland
United States Children's Hospital Birmingham Alabama
United States Roswell Park Cancer Inst Buffalo New York
United States University of North Carolina Chapel Hill North Carolina
United States Carolinas Healthcare System Charleston South Carolina
United States Ann and Robert H Lurie Childrens Hospital of Chicago Chicago Illinois
United States Cincinnati Children's Hospital Medical Center Cincinnati Ohio
United States Nationwide Children's Hospital Columbus Ohio
United States University of Texas Southwestern Medical Center Dallas Texas
United States Golisano Children's Hospital of Southwest Florida Fort Myers Florida
United States Greenville Health System Greenville South Carolina
United States Hackensack University Medical Center Hackensack New Jersey
United States Connecticut Children's Medical Center Hartford Connecticut
United States Penn State Milton S Hershey Medical Center Hershey Pennsylvania
United States Texas Children's Cancer Center Houston Texas
United States Riley Hospital For Children at IU Health Indianapolis Indiana
United States University of Mississippi Medical Center Jackson Mississippi
United States Nemours Children's Clinic Jacksonville Florida
United States Children's Mercy Hospital Kansas City Missouri
United States Children's Specialty Center of Nevada Las Vegas Nevada
United States Arkansas Children's Hospital Little Rock Arkansas
United States Miller Children's Hospital Long Beach California
United States Children's Hospital of Los Angeles Los Angeles California
United States Southern California Permanente Medical Group Los Angeles California
United States Kosair Children's Hospital Louisville Kentucky
United States Valley Children's Hospital Madera California
United States Midwest Children's Cancer Center Milwaukee Wisconsin
United States Children's Hospitals and Clinics of Minnesota Minneapolis Minnesota
United States University of Minnesota Minneapolis Minnesota
United States Morristown Memorial Hosp Morristown New Jersey
United States Vanderbilt University Medical Center Nashville Tennessee
United States Cancer Institute of New Jersey New Brunswick New Jersey
United States Children's Hospital New Orleans New Orleans Louisiana
United States Columbia University Medical Center New York New York
United States Children's Hospital of The King's Daughters Norfolk Virginia
United States Advocate Chilldren's Hospital Oak Lawn Illinois
United States Children's Hospital of Orange County Orange California
United States Lucile Salter Packard Children's Hospital at Stanford Palo Alto California
United States Phoenix Childrens Hospital Phoenix Arizona
United States UPMC Childrens Hospital of Pittsburgh Pittsburgh Pennsylvania
United States Legacy Emanuel Hospital and Health Center Portland Oregon
United States University of Rochester Medical Center Rochester New York
United States Washington University School of Medicine Saint Louis Missouri
United States All Children's Hospital Saint Petersburg Florida
United States Primary Children's Medical Center Salt Lake City Utah
United States Methodist Hospital San Antonio Texas
United States Loma Linda University San Bernardino California
United States UCSF Children's Hospital San Francisco California
United States Seattle Children's Hospital Seattle Washington
United States New York Medical College Valhalla New York
United States Children's Hospital National Medical Center Washington District of Columbia
United States Alfred I Dupont Hospital For Children Wilmington Delaware

Sponsors (1)

Lead Sponsor Collaborator
Celgene

Countries where clinical trial is conducted

United States,  Canada, 

Outcome

Type Measure Description Time frame Safety issue
Primary Number of Participants Who Achieved a Morphologic Complete Response Within the First Four Cycles of Lenalidomide Treatment According to the Modified International Working Group (IWG) Criteria The morphological complete response rate was defined as the total number of participants with morphological CR observed within the first 4 cycles of lenalidomide (regardless of whether the CR/CRi was observed at the end of Cycle 1, 2, 3 or 4) over the total number of participants evaluable for this endpoint. According to Modified IWG criteria, morphologic CR was defined as:
Absolute neutrophil count (ANC) = 1000/µL and platelets = 100,000 without transfusions and/or exogenous growth factor support (i.e., no transfusion or exogenous growth factor within 7 days of assessment;
Bone marrow < 5% blasts evidence of trilineage hematopoiesis;
No evidence of extramedullary disease.
Morphologic CRi was defined as:
ANC < 1000/µL and platelets < 100,000/µL or > 100,000/µL without platelet recovery (requiring transfusion within 7 days of assessment);
BM with < 5% blasts and evidence of trilineage hematopoiesis;
No evidence of extramedullary disease.
From day of the first dose of IP to end of cycle 4; Response was assessed at the completion of the 21-day treatment period of cycles 1, 2, 3, and 4 and at treatment discontinuation.
Secondary Number of Participants Who Achieved a Bone Marrow Confirmed CR/CRi Lasting 3 Months (Durable Response Rate) Durable response rate was defined as the percentage of participants who achieved a BM confirmed CR/CRi according to the Modified IWG Response Assessment Lasting 3 Months (from the time to complete response observed until treatment failure or worse) or until transplantation if earlier among all participants eligible for durable response rate analysis, provided the CR/CRi was confirmed in a bone marrow sample). Due to scarcity of relevant data, it was not practical or meaningful to analyze the durable response rate. Only 1 participant had a response and the participant was censored soon after, as the consent was withdrawn; unable to calculate duration of response. From date of confirmed complete response observed until treatment failure or worse; up to data cut-off date of 31 December 2017
Secondary Duration of Response Duration of response was defined as the time from date of the first observed response (CR, CRi or PR) until morphologic relapse, molecular/cytogenetic relapse, or death only for participants who achieved a response. Due to scarcity of relevant data, it was not practical or meaningful to analyze the duration of response. Only 1 participant had a response and the participant was censored soon after, as the consent was withdrawn; unable to calculate duration of response. From date of first time of complete response observed until treatment failure or worse; up to data cut-off date of 31 December 2017
Secondary Number of Participants Who Achieved a Best Response of Morphologic Complete Remission, Morphologic Complete Remission Incomplete or Partial Remission Overall response rate was defined as the number of participants with best response of CR, CRi or PR.
A CR was defined as:
ANC = 1000/µL and platelets = 100,000 without transfusions and/or exogenous growth factor support (no transfusion or exogenous growth factor within 7 days of assessment);
BM < 5% blasts evidence of trilineage hematopoiesis;
No evidence of extramedullary disease.
A CRi was defined as:
ANC < 1000/µL and platelets < 100,000/µL or > 100,000/µL without platelet recovery (requiring transfusion within 7 days of assessment);
BM with < 5% blasts and evidence of trilineage hematopoiesis;
No evidence of extramedullary disease.
A PR was defined as:
ANC of = 1000/µL and platelets = 100,000 without transfusions and/or exogenous growth factor support (no transfusion or exogenous growth factor within 7 days of assessment);
BM with 5% to 25% blasts and at least a 50% decrease in BM blast percent from baseline;
No evidence of extramedullary disease.
Response was assessed at the completion of the 21-day treatment period of cycles 1, 2, 3.
Secondary Number of Participants With a Morphologic CR, CRi, PR or Treatment Failure at Cycles 1, 2 and 3 Disease assessment outcome at the end of Cycles 1-3 based on Cheson criteria:
Morphologic CR =
ANC = 1000/µL and platelet = 100,000 without transfusions and/or exogenous growth factor support (no transfusion or exogenous growth factor within 7 days of assessment)
BM < 5% blasts evidence of trilineage hematopoiesis
No evidence of extramedullary disease Morphologic CRi =
1. ANC< 1000/µL and Platelets < 100,000/µL or > 100,000/µL without platelet recovery (requiring transfusion within 7 days of assessment) 2. BM with < 5% blasts and evidence of trilineage hematopoiesis 3. No evidence of extramedullary disease PR =
ANC = 1000/µL and platelets = 100,000 without transfusions and/or exogenous growth factor support
BM with < 5%-25% blasts and at least a 50% decrease in BM blast percent from baseline
No evidence of extramedullary disease Treatment Failure = resistant disease; survival = 7 days post-therapy; failed to achieve CR, CRi, or PR but stable with persistent AML
Response was assessed at the completion of the 21-day treatment period of cycles 1, 2, 3.
Secondary Number of Participants Who Received a Haematopoietic Stem Cell Transplant (HSCT) The number of participants who had undergone a haematopoietic stem cell transplant was calculated over the total number of participants in the ITT population. Percentages were also calculated based on whether the transplantation was the first, second, or subsequent transplant post IP administration. From first dose of study drug up to 5 years post HSCT
Secondary Number of Participants Who Experienced Treatment Emergent Adverse Events (TEAE) A TEAE was defined as any adverse event (AE) occurring or worsening on or after the first treatment of lenalidomide and within 28 days after the last dose. A serious AE = any AE which results in death; is life-threatening; requires inpatient hospitalization or prolongation of existing hospitalization; results in persistent or significant disability/incapacity; is a congenital anomaly/birth defect; constitutes an important medical event. The severity of AEs was graded based on National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE), Version 4.3 and based on the following scale: Grade 1 = Mild Grade 2 = Moderate Grade 3 = Severe Grade 4 = Life threatening Grade 5 = Death. From the first dose of study drug until 28 days after the last dose of study drug; up to data cut off date of 31 December 2017; maximum duration of treatment was 12 weeks
Secondary Percentage of Participants With of Graft Versus Host Disease (GVHD) Acute graft versus host disease generally occurs after allogeneic hematopoietic stem cell transplantation. It is a reaction of donor immune cells against host tissues. The 3 main tissues that acute GVHD affects are the skin, liver, and gastrointestinal tract. Chronic GVHD is scored per the National Institute of Health consensus conference grading system. Clinical manifestations of chronic GVHD include skin involvement resembling lichen planus or the cutaneous manifestations of scleroderma; dry oral mucosa with ulcerations and sclerosis of the gastrointestinal tract; and a rising serum bilirubin concentration. From the first dose of study drug to 28 days after the last dose of study drug; up to data cut off date of 31 December 2017; maximum treatment was 12 weeks
Secondary Area Under the Plasma Concentration-time Curve From Time 0 to the Time of the Last Quantifiable Concentration of Lenalidomide (AUC-t) Area under the plasma concentration-time curve from time 0 to the time of the last quantifiable concentration, calculated by linear trapezoidal method when concentrations were increasing and the logarithmic trapezoidal method when concentrations were decreasing. Pharmacokinetic (PK) sampling was conducted after lenalidomide administration at Cycle 1 and was weight dependent at these timepoints: 0.5, 1, 2, 4, 6, 8 and 24 hours. The 24 hour PK sample was taken prior to the lenalidomide dose on Day 2.
Secondary Area Under the Plasma Concentration Time Curve From 0 Extrapolated to Infinity (AUC-inf, AUC08) Of Lenalidomide Area under the plasma concentration-time curve from time 0 extrapolated to infinity, calculated as [AUCt + Ct/ ?z]. Ct is the last quantifiable concentration. No AUC extrapolation was performed with unreliable ?z. Pharmacokinetic sampling was conducted after lenalidomide administration at Cycle 1 and was weight dependent at these timepoints: 0.5, 1, 2, 4, 6, 8 and 24 hours. The 24 hour PK sample was taken prior to the lenalidomide dose on Day 2.
Secondary Maximum Observed Concentration (Cmax) of Lenalidomide Maximum observed plasma concentration, obtained directly from the observed concentration versus time data. PK sampling was conducted after lenalidomide administration at Cycle 1 and was weight dependent at these timepoints: 0.5, 1, 2, 4, 6, 8 and 24 hours. The 24 hour PK sample was taken prior to the lenalidomide dose on Day 2.
Secondary Time to Reach Maximum Concentration (Tmax) of Lenalidomide Time to cmax was obtained directly from the observed concentration versus time data. Pk sampling was conducted after lenalidomide administration at Cycle 1 and was weight dependent at these timepoints: 0.5, 1, 2, 4, 6, 8 and 24 hours. The 24 hour PK sample was taken prior to the lenalidomide dose on Day 2.
Secondary Terminal Half-Life (t1/2) of Lenalidomide Terminal phase half-life in plasma, calculated as [(ln 2)/?z]. Terminal half-life was only calculated when a reliable estimate for ?z could be obtained. Pharmacokinetic sampling was conducted after lenalidomide administration at Cycle 1 and was weight dependent at these timepoints: 0.5, 1, 2, 4, 6, 8 and 24 hours. The 24 hour PK sample was taken prior to the lenalidomide dose on Day 2.
Secondary Apparent Total Clearance (CL/F) of Lenalidomide Apparent volume of distribution, calculated as [(CL/F)/?z]. Pharmacokinetic sampling was conducted after lenalidomide administration at Cycle 1 and was weight dependent at these timepoints: 0.5, 1, 2, 4, 6, 8 and 24 hours. The 24 hour PK sample was taken prior to the lenalidomide dose on Day 2.
Secondary Apparent Volume of Distribution (Vz/F) of Lenalidomide Apparent volume of distribution, calculated as [(CL/F)/?z]. Pharmacokinetic sampling was conducted after lenalidomide administration at Cycle 1 and was weight dependent at these timepoints: 0.5, 1, 2, 4, 6, 8 and 24 hours. The 24 hour PK sample was taken prior to the lenalidomide dose on Day 2.
Secondary Correlation of Peripheral White Blood Cell Count, Absolute Blast Count and Cytogenetics With Response to Lenalidomide Correlation of peripheral white blood cell count, absolute blast count and cytogenetics with response to lenalidomide was not performed since only 1 participant met the primary efficacy endpoint of morphological CR/CRi, and due to scarcity of relevant data, it was not practical or meaningful to analyze to perform the analysis on blood counts and response to lenalidomide. Not Performed
See also
  Status Clinical Trial Phase
Completed NCT02916979 - Myeloid-Derived Suppressor Cells and Checkpoint Immune Regulators' Expression in Allogeneic SCT Using FluBuATG Phase 1
Recruiting NCT04265963 - CD123-Targeted CAR-T Cell Therapy for Relapsed/Refractory Acute Myeloid Leukemia Phase 1/Phase 2
Completed NCT00233909 - A Trial of Gemtuzumab Ozogamicin (GO) in Combination With Zosuquidar in Patients With CD33 Positive Acute Myeloid Leukemia Phase 1/Phase 2
Terminated NCT04748848 - A Safety, Tolerability and Preliminary Efficacy Study of CC-90011 in Combination With Venetoclax and Azacitidine in R/R Acute Myeloid Leukemia and Treatment-naïve Participants Not Eligible for Intensive Therapy Phase 1
Completed NCT04039100 - Family Caregiver Ambassador Support for Family Caregivers of Patients With Hematological Disease N/A
Recruiting NCT03600558 - Efficacy of FLAG Regimen for the Treatment of Patients With AML
Active, not recruiting NCT02333838 - Reduced Toxicity Conditioning Prior to Unrelated Cord Cell Transplantation for High Risk Myeloid Malignancies Phase 2
Completed NCT01061177 - Nilotinib in Newly Diagnosed Adult Philadelphia Chromosome and /or BCR-ABL Positive Chronic Myeloid Leukaemia in Chronic Phase Phase 4
Completed NCT00025662 - Selective T-Cell Depletion to Reduce GVHD (Patients) Receiving Stem Cell Tx to Treat Leukemia, Lymphoma or MDS Phase 2
Terminated NCT02370888 - Lenalidomide After Allo-Hematopoietic Cell Transplant (HCT) in Acute Myelogenous Leukemia (AML) and Myelodysplastic Syndromes (MDS) Subjects With Minimal Residual Disease Phase 1
Recruiting NCT05805605 - Allo HSCT Using RIC and PTCy for Hematological Diseases Phase 2
Recruiting NCT03080922 - High Dose Donor Hematopoietic Stem Cell Infusion for Relapsed/Refractory AML Phase 1/Phase 2
Completed NCT01392989 - Post T-plant Infusion of Allogeneic Cytokine Induced Killer (CIK) Cells as Consolidative Therapy in Myelodysplastic Syndromes/Myeloproliferative Disorders Phase 2
Terminated NCT00306332 - T-cell and B-cell Depletion in Allogeneic Peripheral Blood Stem Cell Transplantation Phase 3
Completed NCT00451997 - Gleevec/Low-Dose Ara-C Study for Elderly Patients With AML and Myelodysplastic Syndromes Phase 2
Recruiting NCT04310592 - Natural Killer Cell (CYNK-001) Infusions in Adults With AML Phase 1
Recruiting NCT05458258 - Studying Malnutrition And Sarcopenia In Older Adults With Acute Myeloid Leukemia
Completed NCT00065143 - Clofarabine Plus Cytarabine in Patients With Previously Untreated Acute Myeloid Leukemia and High-risk Myelodysplastic Syndrome Phase 2
Recruiting NCT04789655 - Study of CC-96191 in Participants With Relapsed or Refractory Acute Myeloid Leukemia Phase 1
Withdrawn NCT04406207 - Endogenous Retroviruses in Acute Myeloid Leukemia