Clinical Trials Logo

Clinical Trial Details — Status: Recruiting

Administrative data

NCT number NCT05457556
Other study ID # ASCT2031
Secondary ID NCI-2022-07080AS
Status Recruiting
Phase Phase 3
First received
Last updated
Start date March 15, 2023
Est. completion date December 1, 2027

Study information

Verified date April 2024
Source Children's Oncology Group
Contact n/a
Is FDA regulated No
Health authority
Study type Interventional

Clinical Trial Summary

This phase III trial compares hematopoietic (stem) cell transplantation (HCT) using mismatched related donors (haploidentical [haplo]) versus matched unrelated donors (MUD) in treating children, adolescents, and young adults with acute leukemia or myelodysplastic syndrome (MDS). HCT is considered standard of care treatment for patients with high-risk acute leukemia and MDS. In HCT, patients are given very high doses of chemotherapy and/or radiation therapy, which is intended to kill cancer cells that may be resistant to more standard doses of chemotherapy; unfortunately, this also destroys the normal cells in the bone marrow, including stem cells. After the treatment, patients must have a healthy supply of stem cells reintroduced or transplanted. The transplanted cells then reestablish the blood cell production process in the bone marrow. The healthy stem cells may come from the blood or bone marrow of a related or unrelated donor. If patients do not have a matched related donor, doctors do not know what the next best donor choice is. This trial may help researchers understand whether a haplo related donor or a MUD HCT for children with acute leukemia or MDS is better or if there is no difference at all.


Description:

PRIMARY OBJECTIVE: I. To compare the 1-year cumulative incidence of severe Graft Versus Host Disease (GVHD) (from day of HCT) defined as grade III-IV acute GVHD (aGVHD) and/or chronic GVHD (cGVHD) that requires systemic immunosuppression and to compare the disease free survival (DFS) (from time of randomization) in children and young adults (AYA) with acute myeloid leukemia (AML), acute lymphoid leukemia (ALL), mixed phenotype acute leukemia (MPAL), and myelodysplastic syndrome (MDS) who are randomly assigned to haploHCT or to an 8/8 adult MUD-HCT. SECONDARY OBJECTIVES: I. To compare overall survival (OS) between children and AYA with AML/ALL/MPAL/MDS randomly assigned to haploHCT and MUD HCT. II. To compare differences in health-related quality of life (HRQOL) between haploHCT and MUD HCT from baseline (pre-transplant), at 6 months, 1 year and 2 years post-transplant. EXPLORATORY OBJECTIVES: I. To compare the median time to engraftment and cumulative incidences of neutrophil engraftment at 30 and 100 days post transplant and platelet engraftment at 60 and 100 days post transplant, primary graft failure by 60 days, secondary graft failure at 1 year post transplant, Grade II-IV and III-IV acute graft versus host disease (aGVHD) requiring systemic immunosuppression at 100 days and 6 months, and cumulative incidences of transplant-related mortality (TRM), relapse, and moderate and severe chronic graft versus host disease (cGVHD) at 6 months, 1 and 2 years after haploHCT and MUD HCT. II. To estimate 1 year, 18-month and 2-year cumulative incidence of graft-versus-host disease (GVHD)-free relapse-free survival (GRFS) with events defined as occurrence of any of the following from Day 0 of HCT: Grade III-IV acute GVHD, chronic GVHD requiring systemic immunosuppressive treatment, disease relapse or progression, and death from any cause. IIa. To compare "chronic GVHD" (GRFS) after haploHCT and MUD HCT using landmark definitions. IIb. To compare "current" GRFS is defined as the time to onset of any of the following events from Day 0 of HCT: Grade III-IV acute GVHD, chronic GVHD that is STILL requiring systemic immunosuppressive treatment, disease relapse or progression, death from any cause at 18 months and 2 years. III. To evaluate the influence of key clinical variables: age (<13 years and 13-21.99 years), disease (ALL/MPAL versus [vs.] AML/MDS), haploHCT approach (TCR alpha beta + T cell depletion vs. post-transplant cyclophosphamide [PTCy]); donor age (by ten-year increments), donor sex (maternal vs. paternal for parental donation), pre-HCT minimal residual disease status (MRD + vs MRD -); pediatric disease risk index (low, intermediate, and high, impact on OS and DFS only), conditioning regimen (chemotherapy based versus total-body irradiation [TBI] based), immunosuppressive regimen (anti-thymocyte globulin [ATG] exposure according to the weight and absolute lymphocyte count [ALC] dependent dosing approach vs no ATG exposure) time to transplant (interval between diagnosis/relapse and date of stem cell infusion) graft cell dose, use of relapse prevention therapy (yes or no) and weight on engraftment, OS, DFS, GRFS, relapse, transplant related mortality (TRM), aGvHD and cGvHD at 1 and 2 years after haplo and MUD HCT by performing stratified and multivariate analyses. IV. To compare other important transplant related outcomes after haplo and MUD HCT, such as: IVa. Incidence of any significant fungal infections (defined as proven or probable fungal infection) through 1 year post HCT; IVb. Incidence of viremia with or without end organ disease (i.e. cytomegalovirus [CMV], adenovirus, Epstein-Barr virus [EBV], human herpesvirus 6 [HHV-6], BK) requiring hospitalization and/or systemic antiviral therapy and/or cell therapy through 1 year post HCT; IVc. Incidence of sinusoidal obstruction syndrome (SOS) through 100 days post HCT; IVd. As defined by the Cairo criteria; IVe. To compare the incidence and outcome of SOS when different criteria are used (European Bone Marrow Transplant [EBMT], Cairo, Baltimore, and modified Seattle criteria); IVf. Incidence of transplant-associated thrombotic microangiopathy (TA-TMA) through 100 days post HCT. V. To compare immune recovery after haplo PTCy, haplo alpha-beta T cell depletion, and MUD HCT via: Va. Pace of reconstitution of T, B, and natural killer (NK) cells and immunoglobulins at 30 days, 60 days, 100 days, 180 days and 365 days after HCT; Vb. Response to vaccinations as determined by vaccination-specific antibody titers at 12-18 months post hematopoietic stem cell transplant (HSCT); Vc. Biobanking blood or marrow to analyze the impact of graft composition on GvHD, relapse and viremia; Vd. Biobanking whole blood and serum to compare immune recovery using extended immune phenotyping and immune functional assessments. VI. Biobanking whole blood or serum to measure rabbit antithymocyte globulin (rATG) exposure when dosed according to weight and absolute lymphocyte count (ALC) using established pharmacokinetic and pharmacodynamics assays (after last infusion, Day -4, Day 0, Day +7). VII. To compare resource utilization after haplo and MUD HCT. VIIa. Length of HCT hospital stay from Day 0 and readmissions within the first 100 days (number of readmissions, duration, and reason). VIIb. Inpatient costs within the first 100 days and at 2 years post HCT. VIII. To describe and compare outcomes (neutrophil and platelet engraftment, graft failure, OS, DFS, GRFS, NRM, relapse, GvHD and health-related quality of life [HRQOL] post HCT) by recipient race/ethnicity, annual household income, primary spoken language and conserved transcriptional response to adversity (CTRA). IX. To describe HRQoL outcomes in racial/ethnic minorities and compare HRQoL outcomes between White patients receiving haploHCT and racial/ethnic minority patients receiving haploHCT. X. To assess the feasibility of incorporating total body irradiation (TBI) delivered with volumetric modulated arc therapy (VMAT) or tomotherapy into a multi-institutional study, to describe the toxicities and oncologic outcomes (relapse, DFS, OS, and TRM) of the subgroup of patients treated with this approach, and to compare these outcomes to those of patients treated with conventional TBI. OUTLINE: Patients who have both a MUD and haplo donor are randomized to Arm A or Arm B. Patients who only have a haplo donor are nonrandomly assigned to Arm C. ARM A: Patients receive a haplo HCT following a TBI- based or chemotherapy-based myeloablative conditioning regimen with PTCy or alpha beta T cell depletion (center's choice). When PTCy is used, it Is administered on days 3 and 4 after HCT and additional immunsouppression is started on day 5 after SCT. ARM B: Patients receive a MUD HCT following a TBI-based or chemotherapy-based myeloablative conditioning regimen between days -9 and -2 Patients then receive GVHD prophylaxis on days 1-11. ARM C: Patients receive a haploHCT following a TBI-based or chemotherapy-based myeloablative conditioning regimen with PTCy or alpha beta T cell depletion (center's choice). When PTCy is used, it Is administered on days 3 and 4 after HCT and additional immunsouppression is started on day 5 after SCT. Patients in all arms undergo standard HCT screening prior to transplant including disease evaluation (lumbar puncture, bone marrow aspiration), and organ function evaluation including but not limited to echocardiogram (ECHO) or multigated acquisition scan (MUGA), PFTS, and bloodwork.Patients also undergo collection of blood throughout the trial. After completion of study treatment, patients are followed periodically for up to 5 years from HCT.


Recruitment information / eligibility

Status Recruiting
Enrollment 435
Est. completion date December 1, 2027
Est. primary completion date December 1, 2027
Accepts healthy volunteers No
Gender All
Age group 6 Months to 21 Years
Eligibility Inclusion Criteria: - PATIENT INCLUSION CRITERIA FOR ENROLLMENT: - 6 months to < 22 years at enrollment - Diagnosed with ALL, AML, or MDS or mixed phenotype acute leukemia (MPAL) for which an allogeneic hematopoietic stem cell transplant is indicated. Complete Remission (CR) status will not be confirmed at the time of enrollment. CR as defined in these sections is required to proceed with the actual HCT treatment plan - Has not received a prior allogeneic hematopoietic stem cell transplant - Does not have a suitable human leukocyte antigen (HLA)-matched sibling donor available for stem cell donation - Has an eligible haploidentical related family donor based on at least intermediate resolution HLA typing - Patients who also have an eligible 8/8 MUD adult donor based on confirmatory high resolution HLA typing are eligible for randomization to Arm A or Arm B. - Patients who do not have an eligible MUD donor are eligible for enrollment to Arm C - All patients and/or their parents or legal guardians must sign a written informed consent - All institutional, Food and Drug Administration (FDA), and National Cancer Institute (NCI) requirements for human studies must be met - Co-Enrollment on other trials - Patients will not be excluded from enrollment on this study if already enrolled on other protocols for treatment of high risk and/or relapsed ALL, AML, MPAL and MDS. This is including, but not limited to, COG AAML1831, COG AALL1821, the EndRAD Trial, as well as local institutional trials. We will collect information on all co-enrollments - Patients will not be excluded from enrollment on this study if receiving immunotherapy prior to transplant as a way to achieve remission and bridge to transplant. This includes chimeric antigen receptor (CAR) T cell therapy and other immunotherapies - PATIENT INCLUSION CRITERIA TO PROCEED TO HCT: - Karnofsky Index or Lansky Play-Performance Scale >= 60 on pre-transplant evaluation. Karnofsky scores must be used for patients >= 16 years of age and Lansky scores for patients =< 16 years of age (within 4 weeks of starting therapy) - A serum creatinine based on age/gender as follows: 6 months to < 1 year: 0.5 mg/dL (Male); 0.5 mg/dL (Female) 1. to < 2 years: 0.6 mg/dL (Male); 0.6 mg/dL (Female) 2. to < 6 years: 0.8 mg/dL (Male); 0.8 mg/dL (Female) 6 to < 10 years: 1 mg/dL (Male); 1 mg/dL (Female) 10 to < 13 years: 1.2 mg/dL (Male); 1.2 mg/dL (Female) 13 to < 16 years: 1.5 mg/dL (Male); 1.4 mg/dL (Female) >= 16 years: 1.7 mg/dL (Male); 1.4 mg/dL (Female) - OR - A 24 hour urine Creatinine clearance >= 60 mL/min/1.73 m^2 - OR - A glomerular filtration rate (GFR) >= 60 mL/min/1.73 m^2. GFR must be performed using direct measurement with a nuclear blood sampling method OR direct small molecule clearance method (iothalamate or other molecule per institutional standard) - Note: Estimated GFR (eGFR) from serum creatinine, cystatin C or other estimates are not acceptable for determining eligibility - Serum glutamic-oxaloacetic transaminase (SGOT) aspartate aminotransferase [AST] or serum glutamate pyruvate transaminase (SGPT) aminotransferase [ALT] < 5 x upper limit of normal (ULN) for age - Total bilirubin < 2.5 mg/dL, unless attributable to Gilbert's Syndrome - Shortening fraction of >= 27% by echocardiogram or radionuclide scan (MUGA) - OR - Ejection fraction of >= 50% by echocardiogram or radionuclide scan (MUGA), choice of test according to local standard of care - Forced expiratory volume in 1 second (FEV1), forced vital capacity (FVC), and corrected carbon monoxide diffusing capability (DLCO) must all be >= 50% of predicted by pulmonary function tests (PFTs). - For children who are unable to perform for PFTs (e.g., due to age or developmental delay), the criteria are: no evidence of dyspnea at rest, oxygen (O2) saturation (Sat) > 92% on room air by pulse oximetry, not on supplemental O2 at rest, and not on supplemental O2 at rest - MPAL in first complete remission (CR1) for whom transplant is indicated. Examples include those patients who are poorly responsive to ALL therapy (end of induction failure( IF-MPAL) to ALL induction (see IF-MPAL note below), end of induction MRD = 5% or end-of-consolidation MRD > 0.01%), as well as patients treated with AML therapy - IF-MPAL: additional criterion for Induction failure for MPAL ONLY as per ALL1732: - An increasing number of circulating leukemia cells on 3 or more consecutive CBCs obtained at daily or longer intervals following day 8 of Induction therapy and prior to day 29 with confirmation by flow cytometry OR development of new sites of extramedullary disease, or other laboratory or clinical evidence of refractory disease or progression prior to the end of Induction evaluation (note that residual testicular disease at the end of Induction is an exception) - MPAL in > second complete remission (CR2) - ALL high-risk in CR1 for whom transplant is indicated. Examples include: induction failure, treatment failure as per minimal residual disease by flow cytometry > 0.01% after consolidation and not eligible for AALL1721 or AALL1721 not available/unwilling to enroll, hypodiploidy (< 44 chromosomes) with MRD+ > 0.01% after induction, persistent or recurrent cytogenetic or molecular evidence of disease during therapy requiring additional therapy after induction to achieve remission (e.g. persistent molecular BCR-ABL positivity), T cell ALL with persistent MRD > 0.01% after consolidation. - ALL in CR2 for whom transplant is indicated. Examples include: B-cell: early (=< 36 months from initiation of therapy) bone marrow (BM) relapse, late BM relapse (>= 36 months) with MRD >= 0.1% by flow cytometry after first re-induction therapy; T or B-cell: early (< 18 months) isolated extramedullary (IEM), late (>= 18 months) IEM, end-Block 1 MRD >= 0.1%; T-cell or Philadelphia chromosome positive (Ph+): BM relapse at any time - ALL in >= third complete remission (CR3) - Patients treated with chimeric antigen receptor T-cells (CART) cells for whom transplant is indicated. Examples include: transplant for consolidation of CART, loss of CART persistence and/or B cell aplasia < 6 months from infusion or have other evidence (e.g., MRD+) that transplant is indicated to prevent relapse - AML in CR1 for whom transplant is indicated. Examples include those deemed high risk for relapse as described in AAML1831: - FLT3/ITD+ with allelic ratio > 0.1 without bZIP CEBPA, NPM1 - FLT3/ITD+ with allelic ratio > 0.1 with concurrent bZIP CEBPA or NPM1 and with evidence of residual AML (MRD >= 0.05%) at end of Induction - Presence of RAM phenotype or unfavorable prognostic markers (other than FLT3/ITD) per cytogenetics, fluorescence in situ hybridization (FISH), next generation sequencing (NGS) results, regardless of favorable genetic markers, MRD status or FLT3/ITD mutation status - AML without favorable or unfavorable cytogenetic or molecular features but with evidence of residual AML (MRD >= 0.05%) at end of Induction - Presence of a non-ITD FLT3 activating mutation and positive MRD (>= 0.05%) at end of Induction 1 regardless of presence of favorable genetic markers. - AML in >= CR2 - MDS with < 5% blasts by morphology and flow cytometry (if available) on the pre-transplant bone marrow evaluation - Complete remission (CR) is defined as < 5% blasts by morphology and flow cytometry (if available) on the pre-transplant bone marrow evaluation with minimum sustained absolute neutrophil count (ANC) of 300 cells/microliter for 1 week or ANC > 500 cells/microliter. We will be collecting data from all approaches to MRD evaluation performed including NGS and polymerase chain reaction (PCR). It is strongly recommended that MPAL be evaluated using multidimensional flow cytometry and/or (KMT2Ar) qt PCR. It is strongly recommended that MPAL be evaluated using multidimensional flow cytometry and/or (KMT2Ar) qt PCR - DONOR ELIGIBILITY CRITERIA: - Matched Unrelated Donors: Unrelated donor candidates must be matched at high resolution at a minimum of 8/8 alleles (HLA-A, -B, -C, -DRB1). One-antigen HLA mismatches are not permitted. HLA matching of additional alleles is recommended according to National Marrow Donor Program (NMDP) guidelines, but will be at the discretion of local centers - Haploidentical Matched Family Members: - Minimum match level full haploidentical (at least 5/10; HLA-A, -B, -C, -DRB1, -DQB1 alleles). The following issues (in no particular order) should be considered in choosing a haploidentical donor: - Absent or low patient donor-specific antibodies (DSA) - Mean fluorescence intensity (MFI) of any anti-donor HLA antibody by solid phase immunoassay should be < 2000. Donors with higher levels are not eligible. - If a screening assay against pooled HLA antigens is used, positive results must be followed with specificity testing using a single antigen assay. The MFI must be < 2000 unless the laboratory has validated higher threshold values for reactivity for HLA antigens (such as HLA-C, -DQ, and -DP), that may be enhanced in concentration on the single antigen assays. Donor anti- recipient antibodies are of unknown clinical significance and do not need to be sent or reported. - Consult with Study Chair for the clinical significance of any recipient anti-donor HLA antibody. - If centers are unable to perform this type of testing, please contact the Study Chair to make arrangements for testing. - If killer immunoglobulin testing (KIR) is performed: KIR status by mismatch, KIR-B, or KIR content criteria can be used according to institutional guidelines. - ABO compatibility (in order of priority): - Compatible or minor ABO incompatibility - Major ABO incompatibility - CMV serostatus: - For a CMV seronegative recipient: the priority is to use a CMV seronegative donor when feasible - For a CMV seropositive recipient: the priority is to use a CMV seropositive donor when feasible - Age: younger donors including siblings/half-siblings, and second degree relatives (aunts, uncles, cousins) are recommended, even if < 18 years - Size and vascular access appropriate by center standard for peripheral blood stem cell (PBSC) collection if needed - Haploidentical matched family members: screened by center health screens and found to be eligible - Unrelated donors: meet eligibility criteria as defined by the NMDP or other unrelated donor registries. If the donor does not meet the registry eligibility criteria but an acceptable eligibility waiver is completed and signed per registry guidelines, the donor will be considered eligible for this study - Human immunodeficiency virus (HIV) negative - Not pregnant - MUD donors and post-transplant cyclophosphamide haplo donors should be asked to provide BM. If donors refuse and other donors are not available, PBSC is allowed. TCR-alpha beta/CD19 depleted haplo donors must agree to donate PBSC - Must give informed consent: - Haploidentical matched family members: Institution standard of care donor consent and Protocol-specific Donor Consent for Optional Studies - Unrelated donors: standard NMDP Unrelated Donor Consent Exclusion Criteria: - PATIENT EXCLUSION CRITERIA FOR ENROLLMENT: - Patients with genetic disorders (generally marrow failure syndromes) prone to secondary AML/ALL/MPAL with known poor outcomes because of sensitivity to alkylator therapy and/or TBI are not eligible (Fanconi Anemia, Kostmann Syndrome, Dyskeratosis Congenita, etc). Patients with Downs syndrome because of increased toxicity with intensive conditioning regimens. - Patients with any obvious contraindication to myeloablative HCT at the time of enrollment - Female patients who are pregnant are ineligible as many of the medications used in this protocol could be harmful to unborn children and infants - Sexually active patients of reproductive potential who have not agreed to use an effective contraceptive method for the duration of their study participation - PATIENT EXCLUSION CRITERIA TO PROCEED TO HCT: - Patients with uncontrolled fungal, bacterial, viral, or parasitic infections are excluded. Patients with history of fungal disease during chemotherapy may proceed if they have a significant response to antifungal therapy with no or minimal evidence of disease remaining by computed tomography (CT) evaluation - Patients with active central nervous system (CNS) leukemia or any other active site of extramedullary disease at the time of initiation of the conditioning regimen are not permitted. - Note: Those with prior history of CNS or extramedullary disease, but with no active disease at the time of pre-transplant workup, are eligible - Pregnant or breastfeeding females are ineligible as many of the medications used in this protocol could be harmful to unborn children and infants

Study Design


Intervention

Procedure:
Biospecimen Collection
Undergo collection of blood
Bone Marrow Aspiration
Undergo bone marrow aspiration
Drug:
Busulfan
Given intravenously (IV)
Cyclophosphamide
Given IV
Procedure:
Echocardiography
Undergo ECHO
Drug:
Fludarabine
Given IV
Procedure:
Haploidentical Hematopoietic Cell Transplantation
Undergo haploHCT
Biological:
Lapine T-Lymphocyte Immune Globulin
Given IV
Procedure:
Lumbar Puncture
Undergo lumbar puncture
Matched Unrelated Donor Hematopoietic Cell Transplantation
Undergo MUD-HCT
Drug:
Melphalan
Given IV
Methotrexate
Given IV
Procedure:
Multigated Acquisition Scan
Undergo MUGA
Drug:
Mycophenolate Mofetil
Given IV
Procedure:
Myeloablative Conditioning
Receive myeloablative conditioning regimen
Other:
Quality-of-Life Assessment
Ancillary studies
Questionnaire Administration
Ancillary studies
Biological:
Rituximab
Given IV
Procedure:
T-Cell Depletion Therapy
Undergo haploHCT with alpha beta T cell depletion
Drug:
Tacrolimus
Given IV
Thiotepa
Given IV
Radiation:
Total-Body Irradiation
Undergo TBI

Locations

Country Name City State
Australia The Children's Hospital at Westmead Westmead New South Wales
Canada Centre Hospitalier Universitaire Sainte-Justine Montreal Quebec
Canada Hospital for Sick Children Toronto Ontario
Canada CancerCare Manitoba Winnipeg Manitoba
United States C S Mott Children's Hospital Ann Arbor Michigan
United States Children's Hospital Colorado Aurora Colorado
United States Johns Hopkins University/Sidney Kimmel Cancer Center Baltimore Maryland
United States Children's Hospital of Alabama Birmingham Alabama
United States Montefiore Medical Center - Moses Campus Bronx New York
United States Roswell Park Cancer Institute Buffalo New York
United States Medical University of South Carolina Charleston South Carolina
United States Lurie Children's Hospital-Chicago Chicago Illinois
United States Cleveland Clinic Foundation Cleveland Ohio
United States Medical City Dallas Hospital Dallas Texas
United States UT Southwestern/Simmons Cancer Center-Dallas Dallas Texas
United States Children's Hospital of Michigan Detroit Michigan
United States City of Hope Comprehensive Cancer Center Duarte California
United States Duke University Medical Center Durham North Carolina
United States Cook Children's Medical Center Fort Worth Texas
United States University of Florida Health Science Center - Gainesville Gainesville Florida
United States Helen DeVos Children's Hospital at Spectrum Health Grand Rapids Michigan
United States Hackensack University Medical Center Hackensack New Jersey
United States Penn State Children's Hospital Hershey Pennsylvania
United States Baylor College of Medicine/Dan L Duncan Comprehensive Cancer Center Houston Texas
United States Riley Hospital for Children Indianapolis Indiana
United States University of Iowa/Holden Comprehensive Cancer Center Iowa City Iowa
United States University of Mississippi Medical Center Jackson Mississippi
United States Nemours Children's Clinic-Jacksonville Jacksonville Florida
United States Children's Mercy Hospitals and Clinics Kansas City Missouri
United States Arkansas Children's Hospital Little Rock Arkansas
United States Loma Linda University Medical Center Loma Linda California
United States Norton Children's Hospital Louisville Kentucky
United States University of Wisconsin Carbone Cancer Center Madison Wisconsin
United States Nicklaus Children's Hospital Miami Florida
United States University of Miami Miller School of Medicine-Sylvester Cancer Center Miami Florida
United States The Children's Hospital at TriStar Centennial Nashville Tennessee
United States Vanderbilt University/Ingram Cancer Center Nashville Tennessee
United States Yale University New Haven Connecticut
United States The Steven and Alexandra Cohen Children's Medical Center of New York New Hyde Park New York
United States Children's Hospital New Orleans New Orleans Louisiana
United States Laura and Isaac Perlmutter Cancer Center at NYU Langone New York New York
United States UCSF Benioff Children's Hospital Oakland Oakland California
United States University of Oklahoma Health Sciences Center Oklahoma City Oklahoma
United States AdventHealth Orlando Orlando Florida
United States Lucile Packard Children's Hospital Stanford University Palo Alto California
United States Phoenix Childrens Hospital Phoenix Arizona
United States Virginia Commonwealth University/Massey Cancer Center Richmond Virginia
United States Mayo Clinic in Rochester Rochester Minnesota
United States University of Rochester Rochester New York
United States Washington University School of Medicine Saint Louis Missouri
United States Johns Hopkins All Children's Hospital Saint Petersburg Florida
United States Primary Children's Hospital Salt Lake City Utah
United States Methodist Children's Hospital of South Texas San Antonio Texas
United States UCSF Medical Center-Mission Bay San Francisco California
United States New York Medical College Valhalla New York
United States Alfred I duPont Hospital for Children Wilmington Delaware

Sponsors (1)

Lead Sponsor Collaborator
Children's Oncology Group

Countries where clinical trial is conducted

United States,  Australia,  Canada, 

Outcome

Type Measure Description Time frame Safety issue
Other Neutrophil engraftment (defined as achieving a donor derived absolute neutrophil count (ANC) >= 500/uL for three consecutive measurements on different days) We will estimate the cumulative incidences of neutrophil engraftment by Day 30 and Day 100 post-HCT and corresponding 95% confidence intervals among enrolled, eligible patients randomized to either the Haplo or MUD arm and who undergo HCT. Day 30 and Day 100 post-transplant
Other Platelet engraftment (defined as the first day of a minimum of three consecutive measurements on different days such that the patient has achieved a platelet count > 20,000/µL and > 50,000/µL with no platelet transfusions in the preceding seven days) We will estimate cumulative incidences of platelet engraftment by Day 30 and Day 100 post-HCT and corresponding 95% confidence intervals among enrolled, eligible patients randomized to either the Haplo or MUD arms and who undergo HCT. Day 30 and Day 100 post-transplant
Other Primary and secondary graft failure Primary (defined as lack of donor-derived neutrophil engraftment) and secondary graft failure (defined as initial donor-derived neutrophil engraftment followed by subsequent decline in ANC to < 500/µL with loss of donor chimerism to < 10% donor CD3 in peripheral blood or bone marrow). We will estimate cumulative incidences of primary and separately, secondary graft failure by Day 60 post-HCT and corresponding 95% confidence intervals among enrolled, eligible patients randomized to either Haplo or MUD arms and who undergo HCT. 60 days post transplant
Other Acute graft versus host disease (aGVHD) Grades II-IV and III-IV We will estimate cumulative incidences of Grade II-IV aGVHD and separately, Grade III-IV aGVHD by Days 100 and 180 post-HCT and corresponding 95% confidence intervals among enrolled, eligible patients randomized to either Haplo or MUD arms and who undergo HCT. Day 100 and Day 180 post-HCT
Other Transplant related mortality (TRM) We will estimate cumulative incidences of transplant-related mortality by 6-months, 1-year, and 2-years post-HCT and corresponding 95% confidence intervals among enrolled, eligible patients randomized to either Haplo or MUD arms and who undergo HCT. 6-months, 1, and 2-year post-HCT
Other Relapse We will estimate cumulative incidences of relapse by 6-months, 1-year, and 2-years post-HCT and corresponding 95% confidence intervals among enrolled, eligible patients randomized to either Haplo or MUD arms. 6-months, 1, and 2-years post-HCT
Other Graft-versus-host disease (GVHD)-free relapse-free survival (GRFS) (using the standard definition and two landmark definitions) We will estimate cumulative incidences of GRFS events using all three definitions by 1-year, 18-months, and 2-years post-HCT and corresponding 95% confidence intervals among enrolled, eligible patients randomized to either Haplo or MUD arms and who undergo HCT. 1-year, 18-months, and 2-years post-HCT
Other Mild, moderate, and severe chronic graft versus host disease (cGVHD) We will estimate cumulative incidences of mild, moderate, and severe cGvHD by 6-months, 1-year, and 2-years post-HCT and corresponding 95% confidence intervals among enrolled, eligible patients randomized to either Haplo or MUD arms and who undergo HCT. 6-months, 1, and 2-year post-HCT
Other Significant fungal infections (defined as proven or probable fungal infection according to the De-Pauw definition) We will estimate the cumulative incidence of significant fungal infections by 1-year post-HCT and the corresponding 95% confidence interval among enrolled, eligible patients randomized to either Haplo or MUD arms. Up-to 1-year post-HCT
Other Viremia (with or without end organ disease) requiring hospitalization and/or systemic antiviral therapy We will estimate the cumulative incidence of viremia by 1-year post-HCT and the corresponding 95% confidence interval among enrolled, eligible patients randomized to either Haplo or MUD arms. Up to 1-year post-HCT
Other Sinusoidal obstruction syndrome (SOS) (defined by the Cairo criteria and, separately, the EBMT, Baltimore, and modified Seattle criteria) We will estimate the proportion of patients experiencing SOS under the separate definitions by 100-days post-HCT and the corresponding 95% confidence interval among enrolled, eligible patients randomized to either Haplo or MUD arms. Up to 100-days post-HCT
Other Transplant-associated thrombotic microangiopathy (TA-TMA) We will estimate the proportion of TA-TMA events up-to 100-days post-HCT and the corresponding 95% confidence interval among enrolled, eligible patients randomized to either Haplo or MUD arms and who undergo HCT. Up to 100-days post-HCT
Other Reconstitution of T, B, and natural killer (NK) cells and immunoglobulins We will estimate the median of the distribution of T, B, NK cells, and immunoglobulins among patients given each of three HCT methods (Haplo PTCy, Haplo alpha-beta T-cell depletion, and MUD). At 30-days, 60-days, 100-days, 180-days and 365-days after HCT
Other Vaccination-specific antibody titers We will estimate theedian antibody titer among patients given each of three HCT methods (haplo PTCy, haplo alpha-beta T-cell depletion, and MUD). 12-18 months post-HSCT
Other Resource utilization (defined using the length of HCT hospital stay including readmissions within 100-days post-HCT, and using costs (inpatient) within the first 100-days and 2-years post HCT using Public Health Information System) The average proportion of days spent hospitalized will be computed for patients randomized to the Haplo and MUD arms and corresponding 95% confidence intervals will be reported. The median inpatient PHIS adjusted costs by day 100 and 2-years post-HCT will be calculated for patients randomized to Haplo and MUD arms. 100-days and 2-years post-HCT
Other incorporating total body irradiation (TBI) delivered with volumetric modulated arc therapy (VMAT) or tomotherapy Will be assessed by quality assurance monitoring. Will review treatment plans along with treatment received using data submitted to IROC and will describe the proportion of patients whose planned and actual TBI treatments align among those who received VMAT or tomotherapy. Will quantify the proportion of patients receiving VMAT or tomotherapy TBI who experience serious adverse events (defined as any AE reported through CTEP-AERS) and, separately, who experience any unexpected AE. Will estimate the cumulative incidence of death or relapse (DFS as a composite event) as well as death and relapse separately, and TRM by 0.5, 1, and 2-years year post-transplant from the time of starting TBI among those who receive VMAT or tomotherapy. For relapse and TRM, competing risk analysis will be used, as described under 9.3.5.1. 95% confidence intervals for each of these point estimates will also be provided. Date of initiating TBI through 100 days post-transplant.
Primary Severe GVHD (Grade III-IV acute GVHD or chronic GVHD requiring systemic immunosuppressive therapy) We will estimate the cumulative incidence of severe GVHD at 1-year post-HCT and corresponding 95% confidence interval among enrolled and eligible patients randomly assigned to either HAPLO or MUD arms who actually undergo HCT. Up-to 1-year post hematopoietic cell transplantation (HCT)
Primary Disease free survival (DFS) (where an event is the occurrence of death from any cause or relapse) We will estimate the cumulative incidence at 1 year post randomization and the corresponding 95% confidence interval among all enrolled and eligible patients randomly assigned to either HAPLO or MUD arms. From date of randomization to up-to 1 year post randomization
Secondary Overall survival (OS) We will estimate the cumulative incidence of all-cause mortality events up-to 1-year post-HCT and the corresponding 95% confidence interval among all enrolled, eligible, randomized patients. From date of randomization to up-to 1-year post-HCT
Secondary Summary score from the Generic Pediatric Quality of Life Inventory (PedsQL) (excluding School Functioning) For enrolled, eligible patients who consent to optional QoL studies, we will estimate the mean PedsQL score (excluding School Functioning) among patients randomized to the Haplo arm, the MUD arm, and among patients who enroll to arm C. For each, we will also calculate a corresponding 95% confidence interval. At 6-months, 1 year and 2 years post-HCT
Secondary Summary score from the PedsQL Stem Cell Transplant module For enrolled, eligible patients who consent to optional QoL studies, we will estimate the mean PedsQL Stem Cell Transplant module score among patients randomized to the Haplo arm and the MUD arm, and among patients who enroll to arm C. For each, we will calculate a corresponding 95% confidence interval. 6-months, 1 year and 2 years post-HCT
See also
  Status Clinical Trial Phase
Suspended NCT05400122 - Natural Killer (NK) Cells in Combination With Interleukin-2 (IL-2) and Transforming Growth Factor Beta (TGFbeta) Receptor I Inhibitor Vactosertib in Cancer Phase 1
Recruiting NCT04460235 - Immunogenicity of an Anti-pneumococcal Combined Vaccination in Acute Leukemia or Lymphoma Phase 4
Active, not recruiting NCT03678493 - A Study of FMT in Patients With AML Allo HSCT in Recipients Phase 2
Completed NCT04022785 - PLX51107 and Azacitidine in Treating Patients With Acute Myeloid Leukemia or Myelodysplastic Syndrome Phase 1
Recruiting NCT05424562 - A Study to Assess Change in Disease State in Adult Participants With Acute Myeloid Leukemia (AML) Ineligible for Intensive Chemotherapy Receiving Oral Venetoclax Tablets in Canada
Completed NCT03197714 - Clinical Trial of OPB-111077 in Patients With Relapsed or Refractory Acute Myeloid Leukaemia Phase 1
Terminated NCT03224819 - Study of Emerfetamab (AMG 673) in Adults With Relapsed/Refractory Acute Myeloid Leukemia (AML) Early Phase 1
Active, not recruiting NCT04070768 - Study of the Safety and Efficacy of Gemtuzumab Ozogamicin (GO) and Venetoclax in Patients With Relapsed or Refractory CD33+ Acute Myeloid Leukemia:Big Ten Cancer Research Consortium BTCRC-AML17-113 Phase 1
Active, not recruiting NCT03844048 - An Extension Study of Venetoclax for Subjects Who Have Completed a Prior Venetoclax Clinical Trial Phase 3
Active, not recruiting NCT04107727 - Trial to Compare Efficacy and Safety of Chemotherapy/Quizartinib vs Chemotherapy/Placebo in Adults FMS-like Tyrosine Kinase 3 (FLT3) Wild-type Acute Myeloid Leukemia (AML) Phase 2
Recruiting NCT04920500 - Bioequivalence of Daunorubicin Cytarabine Liposomes in Naive AML Patients N/A
Recruiting NCT04385290 - Combination of Midostaurin and Gemtuzumab Ozogamicin in First-line Standard Therapy for Acute Myeloid Leukemia (MOSAIC) Phase 1/Phase 2
Recruiting NCT03897127 - Study of Standard Intensive Chemotherapy Versus Intensive Chemotherapy With CPX-351 in Adult Patients With Newly Diagnosed AML and Intermediate- or Adverse Genetics Phase 3
Active, not recruiting NCT04021368 - RVU120 in Patients With Acute Myeloid Leukemia or High-risk Myelodysplastic Syndrome Phase 1
Recruiting NCT03665480 - The Effect of G-CSF on MRD After Induction Therapy in Newly Diagnosed AML Phase 2/Phase 3
Completed NCT02485535 - Selinexor in Treating Patients With Intermediate- and High-Risk Acute Myeloid Leukemia or High-Risk Myelodysplastic Syndrome After Transplant Phase 1
Enrolling by invitation NCT04093570 - A Study for Participants Who Participated in Prior Clinical Studies of ASTX727 (Standard Dose), With a Food Effect Substudy at Select Study Centers Phase 2
Recruiting NCT04069208 - IA14 Induction in Young Acute Myeloid Leukemia Phase 2
Recruiting NCT05744739 - Tomivosertib in Relapsed or Refractory Acute Myeloid Leukemia (AML) Phase 1
Recruiting NCT04969601 - Anti-Covid-19 Vaccine in Children With Acute Leukemia and Their Siblings Phase 1/Phase 2