Mismatched Related Donor Versus Matched Unrelated Donor Stem Cell Transplantation for Children, Adolescents, and Young Adults With Acute Leukemia or Myelodysplastic Syndrome

T
Toshihiro Onishi, MD

Primary Investigator

Overview

This phase III trial compares hematopoietic (stem) cell transplantation (HCT) usinghed related donors (haploidentical [haplo]) versus matched unrelated donors (MUD) ing children, adolescents, and young adults with acute leukemia or myelodysplasticyndrome (MDS). HCT is considered standard of care treatment for patients with high-riskute leukemia and MDS. In HCT, patients are given very high doses of chemotherapy ordiation therapy, which is intended to kill cancer cells that may be resistant to moredard 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 supplyduced or transplanted. The transplanted cells then reestablish the bloodduction 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 or if a haplo related donor or MUD is better. This trial may help researchers understand whether a haplo related donor or a MUD HCThildren 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) thatquires systemic immunosuppression and to compare the disease free survival (DFS) (from timedomization) in children and young adults (AYA) with acute myeloid leukemia (AML), acuteymphoid leukemia (ALL), 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/MDS randomlygned 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 neutrophilgraftment at 30 and 100 days post transplant and platelet engraftment at 60 and 100 daysy graft failure by 60 days, secondary graft failure at 1 year postGrade II-IV and III-IV acute graft versus host disease (aGVHD) requiring systemicunosuppression at 100 days and 6 months, and cumulative incidences of transplant-relatedy (TRM), relapse, and moderate and severe chronic graft versus host disease (cGVHD)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 thewing from Day 0 of HCT: Grade III-IV acute GVHD, chronic GVHD requiring systemicunosuppressive 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 eventsDay 0 of HCT: Grade III-IV acute GVHD, chronic GVHD that is STILL requiring systemicunosuppressive treatment, disease relapse or progression, death from any cause at 18hs and 2 years.
III. To evaluate the influence of key clinical variables: age (<13 years and 13-21.99 years), disease (ALL versus [vs.] AML/MDS), haploHCT approach (TCR alpha beta + T cell depletion vs.yclophosphamide [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 DFSy), conditioning regimen (chemotherapy based versus total-body irradiation [TBI] based),unosuppressive regimen (anti-thymocyte globulin [ATG] exposure according to the weight andbsolute lymphocyte count [ALC] dependent dosing approach vs no ATG exposure) time to(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,d mortality (TRM), aGvHD and cGvHD at 1 and 2 years after haplo andD 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) 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 cellherapy through 1 year post HCT; IVc. Incidence of sinusoidal obstruction syndrome (SOS)hrough 100 days post HCT; IVd. As defined by the Cairo criteria; IVe. To compare thedence and outcome of SOS when different criteria are used (European Bone Marrow Transplant [EBMT], Cairo, Baltimore, and modified Seattle criteria); IVf. Incidence ofd 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(HSCT); Vc. Biobanking blood or marrow to analyze the impact of graftGvHD, relapse and viremia; Vd. Biobanking whole blood and serum to compareune 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 establishedharmacokinetic 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 hospitaly from Day 0 and readmissions within the first 100 days (number of readmissions, duration,d reason).
VIIb. Inpatient costs within the first 100 days and at 2 years post HCT. VIII. To described compare outcomes (neutrophil and platelet engraftment, graft failure, OS, DFS, GRFS, NRM,GvHD and health-related quality of life [HRQOL] post HCT) by recipient/ethnicity, area-based socioeconomic (SES) status, annual household income, primaryken 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.
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-basedyeloablative conditioning regimen with PTCy or alpha beta T cell depletion (center'shoice). When PTCy is used, it Is administered on days 3 and 4 after HCT and additionalunsouppression is started on day 5 after SCT.
ARM B: Patients receive a MUD HCT following a TBI-based or chemotherapy-based myeloablativeditioning 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 myeloablativeditioning regimen with PTCy or T cell depletion (center's choice). When PTCy is used, itdministered 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 diseasevaluation (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 yearsHCT.

Eligibility

You may be eligible for this study if you meet the following criteria:

  • Conditions:
    Acute Lymphoblastic Leukemia, Acute Myeloid Leukemia, Myelodysplastic Syndrome
  • Age: Between 6 Months - 21 Years
  • Gender: All

Inclusion Criteria:
  • PATIENT INCLUSION CRITERIA FOR ENROLLMENT:
  • 6 months to < 22 years at enrollment
  • Diagnosed with ALL, AML, or MDS for which an allogeneic hematopoietic stem celldicated. Complete Remission (CR) status will not be confirmed at the. CR as defined in these sections is required to proceed with theual 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 availabledonation
  • Has an eligible haploidentical related family donor based on at least intermediateution HLA typing
    • Patients who also have an eligible 8/8 MUD adult donor based on confirmatory highution 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 ArmC
  • 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 enrolledher protocols for treatment of high risk and/or relapsed ALL, AML and MDS.This is including, but not limited to, COG AAML1831, COG AALL1821, the EndRADTrial, as well as local institutional trials. We will collect information on all
    • Patients will not be excluded from enrollment on this study if receivingunotherapy prior to transplant as a way to achieve remission and bridge to. This includes chimeric antigen receptor (CAR) T cell therapy andher 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=< 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)RA 24 hour urine Creatinine clearance >= 60 mL/min/1.73 m^2RA glomerular filtration rate (GFR) >= 60 mL/min/1.73 m^2. GFR must be performed usingdirect measurement with a nuclear blood sampling method OR direct small moleculehod (iothalamate or other molecule per institutional standard): Estimated GFR (eGFR) from serum creatinine, cystatin C or other estimatesble for determining eligibilityum glutamic-oxaloacetic transaminase (SGOT) aspartate aminotransferase [AST] orum glutamate pyruvate transaminase (SGPT) aminotransferase [ALT] < 5 x upper limit(ULN) for ageTotal bilirubin < 2.5 mg/dL, unless attributable to Gilbert's Syndromehortening fraction of >= 27% by echocardiogram or radionuclide scan (MUGA)Rjection fraction of >= 50% by echocardiogram or radionuclide scan (MUGA), choice ofding to local standard of careForced expiratory volume in 1 second (FEV1), forced vital capacity (FVC), andd carbon monoxide diffusing capability (DLCO) must all be >= 50% of predictedby pulmonary function tests (PFTs).For children who are unable to perform for PFTs (e.g., due to age ordevelopmental delay), the criteria are: no evidence of dyspnea at rest, oxygen(O2) saturation (Sat) > 92% on room air by pulse oximetry, not on supplemental O2d not on supplemental O2 at restALL high-risk in first complete remission (CR1) for whom transplant is indicated.ude: induction failure, treatment failure as per minimal residual diseaseby flow cytometry > 0.01% after consolidation and not eligible for AALL1721 orAALL1721 not available/unwilling to enroll, hypodiploidy (< 44 chromosomes) with MRD+> 0.01% after induction, persistent or recurrent cytogenetic or molecular evidence ofdisease during therapy requiring additional therapy after induction to achieve(e.g. persistent molecular BCR-ABL positivity), T cell ALL with persistentRD > 0.01% after consolidation.ALL in second complete remission (CR2) for whom transplant is indicated. Examplesude: B-cell: early (=< 36 months from initiation of therapy) bone marrow (BM)BM relapse (>= 36 months) with MRD >= 0.1% by flow cytometry after firstduction therapy; T or B-cell: early (< 18 months) isolated extramedullary (IEM),(>= 18 months) IEM, end-Block 1 MRD >= 0.1%; T-cell or Philadelphia chromosomeve (Ph+): BM relapse at any timeALL in >= third complete remission (CR3)Patients treated with chimeric antigen receptor T-cells (CART) cells for whomdicated. Examples include: transplant for consolidation of CART, lossCART persistence and/or B cell aplasia < 6 months from infusion or have othervidence (e.g., MRD+) that transplant is indicated to prevent relapseAML in CR1 for whom transplant is indicated. Examples include those deemed high riskdescribed in AAML1831:FLT3/ITD+ with allelic ratio > 0.1 without bZIP CEBPA, NPM1FLT3/ITD+ with allelic ratio > 0.1 with concurrent bZIP CEBPA or NPM1 and withvidence of residual AML (MRD >= 0.05%) at end of InductionPresence of RAM phenotype or unfavorable prognostic markers (other than FLT3/ITD)ytogenetics, fluorescence in situ hybridization (FISH), next generationquencing (NGS) results, regardless of favorable genetic markers, MRD status orFLT3/ITD mutation statusAML without favorable or unfavorable cytogenetic or molecular features but withvidence of residual AML (MRD >= 0.05%) at end of InductionPresence of a non-ITD FLT3 activating mutation and positive MRD (>= 0.05%) at endduction 1 regardless of presence of favorable genetic markers.AML in >= CR2DS with < 5% blasts by morphology and flow cytometry (if available) on thebone marrow evaluationComplete remission (CR) is defined as < 5% blasts by morphology and flow cytometry (ifvailable) on the pre-transplant bone marrow evaluation with minimum sustainedbsolute neutrophil count (ANC) of 300 cells/microliter for 1 week or ANC > 500/microliter. We will be collecting data from all approaches to MRD evaluationd including NGS and polymerase chain reaction (PCR)DONOR ELIGIBILITY CRITERIA:hed Unrelated Donors:d 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 ofdditional alleles is recommended according to National Marrow Donor Program (NMDP)guidelines, but will be at the discretion of local centersHaploidentical Matched Family Members:um match level full haploidentical (at least 5/10; HLA-A, -B, -C, -DRB1,DQB1 alleles). The following issues (in no particular order) should bedered in choosing a haploidentical donor:Absent or low patient donor-specific antibodies (DSA)uorescence intensity (MFI) of any anti-donor HLA antibody byd phase immunoassay should be < 2000. Donors with higher levels aregible.g assay against pooled HLA antigens is used, positiveults must be followed with specificity testing using a singlegen assay. The MFI must be < 2000 unless the laboratory hasvalidated higher threshold values for reactivity for HLA antigens(such as HLA-C, -DQ, and -DP), that may be enhanced inhe single antigen assays. Donor anti- recipientbodies are of unknown clinical significance and do not need tobe sent or reported.Consult with Study Chair for the clinical significance of anydonor HLA antibody.unable to perform this type of testing, pleasehe Study Chair to make arrangements for testing.killer immunoglobulin testing (KIR) is performed: KIR status by mismatch,KIR-B, or KIR content criteria can be used according to institutionalguidelines.ABO compatibility (in order of priority):Compatible or minor ABO incompatibilityjor ABO incompatibilityCMV serostatus:For a CMV seronegative recipient: the priority is to use a CMVgative donor when feasibleFor a CMV seropositive recipient: the priority is to use a CMVve donor when feasibleAge: younger donors including siblings/half-siblings, and second degreeves (aunts, uncles, cousins) are recommended, even if < 18 yearsze and vascular access appropriate by center standard for peripheral blood stem cell(PBSC) collection if neededHaploidentical matched family members: screened by center health screens and found tobe eligibled donors: meet eligibility criteria as defined by the NMDP or other unrelateddonor registries. If the donor does not meet the registry eligibility criteria but anble eligibility waiver is completed and signed per registry guidelines, thedonor will be considered eligible for this studyHuman immunodeficiency virus (HIV) negativegnantD donors and post-transplant cyclophosphamide haplo donors should be asked tovide BM. If donors refuse and other donors are not available, PBSC is allowed.TCR-alpha beta/CD19 depleted haplo donors must agree to donate PBSCust give informed consent:Haploidentical matched family members: Institution standard of care donor consentd Protocol-specific Donor Consent for Optional Studiesd donors: standard NMDP Unrelated Donor Consentusion Criteria:PATIENT EXCLUSION CRITERIA FOR ENROLLMENT:Patients with genetic disorders (generally marrow failure syndromes) prone todary AML/ALL with known poor outcomes because of sensitivity to alkylator therapyd/or TBI are not eligible (Fanconi Anemia, Kostmann Syndrome, DyskeratosisCongenita, etc). Patients with Downs syndrome because of increased toxicity withve conditioning regimens.Patients with any obvious contraindication to myeloablative HCT at the time ofFemale patients who are pregnant are ineligible as many of the medications used inhis protocol could be harmful to unborn children and infantsually active patients of reproductive potential who have not agreed to use anve contraceptive method for the duration of their study participationPATIENT EXCLUSION CRITERIA TO PROCEED TO HCT:Patients with uncontrolled fungal, bacterial, viral, or parasitic infections areuded. Patients with history of fungal disease during chemotherapy may proceed ifhey have a significant response to antifungal therapy with no or minimal evidence ofdisease remaining by computed tomography (CT) evaluationPatients with active central nervous system (CNS) leukemia or any other active site ofdullary disease at the time of initiation of the conditioning regimen are notd.: Those with prior history of CNS or extramedullary disease, but with nove disease at the time of pre-transplant workup, are eligiblePregnant or breastfeeding females are ineligible as many of the medications used inhis protocol could be harmful to unborn children and infants

Updated on 27 Apr 2024. Study ID: ASCT2031
Please visit our main page to search for other studies you may be interested in. If you need help finding a study or have any questions, please contact us at inhealth@iu.edu

Interested in the study?

Select a study center that’s convenient for you, and get in touch with the study team.

Contact a study center