Haploidentical Hematopoietic Stem Cell Transplantation With Ex Vivo TCR Alpha/Beta and CD19 Depletion in Pediatric Hematologic Malignancies

A Pilot Study of Haploidentical Hematopoietic Stem Cell Transplantation With Ex Vivo TCR Alpha/Beta and CD19 Depletion in Pediatric Hematologic Malignancies

This single arm pilot phase I study with safety run-in is designed to estimate the safety and efficacy of a familial mismatched or haploidentical hematopoietic stem cell transplantation (haplo-HSCT) using a novel graft modification technique (selective αβ-TCR and CD19 depletion).

Study Overview

Study Type

Interventional

Enrollment (Estimated)

32

Phase

  • Phase 1

Contacts and Locations

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

Study Contact

Study Locations

    • Missouri
      • Saint Louis, Missouri, United States, 63110
        • Recruiting
        • Washington University School of Medicine
        • Contact:
        • Sub-Investigator:
          • Megha Malhotra, M.D.
        • Sub-Investigator:
          • Rachel Langley, PharmD
        • Sub-Investigator:
          • Thomas Pfeiffer, M.D.
        • Sub-Investigator:
          • Melissa Mavers, M.D., Ph.D.
        • Sub-Investigator:
          • Feng Gao, Ph.D.
        • Sub-Investigator:
          • Shalini Shenoy, M.D.
        • Sub-Investigator:
          • Robert Hayashi, M.D.
        • Principal Investigator:
          • Jeffrey Bednarski, M.D., Ph.D.

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

No older than 30 years (Child, Adult)

Accepts Healthy Volunteers

No

Description

Recipient Inclusion Criteria:

  • Must meet at least one of the following disease criteria:

    • B cell ALL in first remission and any of the following:

      • Persistent flow-based MRD at end-of-consolidation:

        • >= 1% for NCI SR ALL
        • >= 0.01% for NCI HR ALL
      • TCF3-HLF t(17;19)
      • KMT2A rearranged infant ALL, < 6 months of age and presenting WBC of > 300,000 or poor steroid response (peripheral blasts >= 1000 /uL on day 8 of therapy
      • Other high-risk features not explicitly stated here, after discussion/approval with protocol PI.
    • B cell ALL in second remission and any of the following:

      • Early (<36 months from start of therapy) marrow or combined relapse
      • Late (>36 months from start of therapy) marrow or combined relapse with end-of re-induction flow MRD >= 0.1%
      • Early isolated extramedullary relapse (< 18 months from start of therapy)
    • Any B cell ALL in third or greater remission
    • T cell ALL in first remission

      • End-of consolidation MRD > 0.1%
    • Any T cell ALL in second or greater remission
    • AML in first remission with any of the following high-risk features:

      • MRD >= 1% after first induction course
      • MRD >= 0.1% after second induction course
      • RPN1-MECOM
      • RUNX1-MECOM
      • NPM1-MLF1
      • DEK-NUP214
      • KAT6A-CREBBP (if >= 90 days at diagnosis)
      • FUS-ERG
      • KMT2A-AFF1
      • KMT2A-AFDN
      • KMT2A-ABI1
      • KMT2A-MLLT1
      • 11p15 rearrangement (NUP98 - any partner gene)
      • 12p13.2 rearrangement (ETV6 - any partner gene)
      • Deletion 12p to include 12p13.2 (loss of ETV6)
      • Monosomy 5/Del(5q) to include 5q31 (loss of EGR1)
      • Monosomy 7
      • 10p12.3 rearrangement (MLLT10b - any partner gene)
      • FLT3/ITD with allelic ratio > 0.1%
      • RAM phenotype as evidenced by flow cytometry: bright CD56+, dim to negative CD45 and CD38 and lack of HLA-DR
      • Other high-risk features not explicitly stated here, after discussion/approval with protocol PI.
    • AML in second or greater remission
    • Mixed phenotype or undifferentiated leukemia in any CR
    • Secondary to therapy-associated leukemia in any CR
    • NK cell lineage leukemia in any CR
    • Myelodysplastic syndrome (MDS)
    • Juvenile myelomonocytic leukemia (JMML)
  • May have undergone a prior hematopoietic stem cell transplant provided one of the criteria in Inclusion Criterion #1 are met AND the patient does not have active GVHD (has been off immunosuppression for at least 3 months).
  • Available familial haploidentical donor.
  • Donor and recipient must be identical at a minimum of one allele of each of the following genetic loci: HLA-A, HLA-B, HLA-Cw, HLA-DRB1, and HLA-DQB1. A minimum of 5/10 match is required and will be considered sufficient evidence that the donor and recipient share one HLA haplotype.
  • No more than 30 years of age
  • Lansky or Karnofsky performance status > 50%
  • Adequate organ function as defined below:

    • Cardiac: LVEF ≥ 40% at rest or SF ≥ 26%
    • Hepatic:

      • Total bilirubin < 3 x IULN for age
      • AST(SGOT)/ALT(SGPT) < 5 x IULN
    • Renal: GFR ≥ 60 mL/min/1.73m2 as estimated by updated Schwartz formula for ages 1-17 years (see Appendix B), 24-hour creatinine clearance, or renal scintigraphy. If GFR is abnormal for age based on updated Schwartz formula, accurate measurement should be obtained by either 24-hour creatinine clearance or renal scintigraphy. Renal function may also be estimated by serum creatinine based on age/gender. A minimum serum creatinine of 2x upper limit of normal is required for inclusion on this protocol.
    • Pulmonary:

      • O2 saturation ≥ 92% on room air without positive pressure support
      • FEV1, FVC, and DLCO ≥ 50% of predicted (for children unable to perform a pulmonary function test, a high-resolution CT chest may be obtained)
  • The effects of these treatments on the developing human fetus are unknown. For this reason, patients of childbearing potential must agree to use adequate contraception (hormonal or barrier method of birth control, abstinence) prior to study entry and for 24 months following transplant. Should a woman become pregnant or suspect she is pregnant while participating in this study, she must inform her treating physician immediately.
  • Ability to understand and willingness to sign an IRB approved written informed consent document (or that of legally authorized representative, if applicable).

Recipient Exclusion Criteria:

  • Available matched related donor. A patient with a matched unrelated donor is eligible if urgent transplantation is required. A prior unrelated donor search is not required for enrollment.
  • Active non-hematologic malignancy. History of other malignancy is acceptable as long as therapy has been complete and there is no evidence of disease.
  • Currently receiving any other investigational agents.
  • Active CNS or extramedullary disease. History of CNS or extramedullary disease now in remission is acceptable.
  • A history of allergic reactions attributed to compounds of similar chemical or biologic composition to conditioning agents used in the study.
  • Uncontrolled intercurrent illness including, but not limited to, ongoing or active infection (bacterial, viral with clinical instability, or fungal), symptomatic congestive heart failure, or unstable cardiac arrhythmia.
  • Presence of significant anti-donor HLA antibodies per institutional standards. Anti-donor HLA Antibody Testing is defined as a positive crossmatch test of any titer (by complement dependent cytotoxicity or flow cytometric testing) or the mean fluorescence intensity (MFI) of any anti-donor HLA antibody by solid phase immunoassay.
  • Presence of a second major disorder deemed a contraindication for HSCT.
  • Pregnant and/or breastfeeding. Women of childbearing potential must have a negative pregnancy test within 14 days of the start of conditioning.

Donor Eligibility Criteria:

  • At least 6 months of age
  • Meets the selection criteria as defined by the Foundation for the Accreditation of Hematopoietic Cell Therapy (FACT).
  • Able to understand and willing to sign an IRB-approved written informed consent document (or that of legally authorized representative, if applicable).

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: N/A
  • Interventional Model: Single Group Assignment
  • Masking: None (Open Label)

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
Experimental: ex vivo αβ-TCR/CD19 depleted haplo-hematopoietic stem cell infusion (HSCT)
  • Patients will undergo standard of care conditioning regiment prior to HSCT
  • On Day 0, patients will undergo infusion of the ex vivo αβ-TCR/CD19 depleted haplo-HSCT from a stimulated peripheral stem cell source per institutional standard of care. Patients whose graft has a residual CD20+ count > 1.0 x 10^5 may receive a single infusion of rituximab on Day +1 at a dose of 375 mg/m^2 at provider's discretion.
Once pheresed, the product will be washed to remove platelets and the cell concentration will be adjusted per laboratory and ClinicMACS technology recommendations. It is then labeled using the CliniMACS αβ-TCR Biotin Kit and CD19+ immunomagnetic microbeads. After labeling, the cells are washed to remove unbound microbeads. The partially processed product is loaded on the CliniMACS device where labeled cells are depleted and the negative fraction is eluted off the device. The negative fraction is centrifuged and volume reconstituted to obtain the final product

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Safety as measured by the number of events occurring within the first 100 days post-transplant
Time Frame: Through 100 days post-transplant
-Events are death, disease recurrence or progression, and graft failure
Through 100 days post-transplant
Engraftment as measured by time to neutrophil count recovery
Time Frame: From day of transplant (day 0) to 42 days (+/- 14 days) post transplant
Time to neutrophil recovery is defined as the first of 3 measurements on different days when the patient has an absolute neutrophil count of >500/μL after conditioning.
From day of transplant (day 0) to 42 days (+/- 14 days) post transplant
Engraftment as measured by time to platelet count recovery
Time Frame: From day of transplant (day 0) to 75 days (+/- 14 days) post transplant)
Time to platelet recovery is defined as the first day of a minimum of 3 measurements on different days that the patient has achieved a platelet count > 50,000/μL AND did not receive a platelet transfusion in the previous 7 days. The exception is the case in which a patient receives platelet transfusions specifically to achieve a higher platelet threshold to allow for an invasive procedure or protection if determined to be at elevated bleeding risk.
From day of transplant (day 0) to 75 days (+/- 14 days) post transplant)
Donor cell chimerism as measured by short tandem repeat analysis
Time Frame: Through day +100
  • Can use peripheral blood samples or bone marrow samples
  • The percent of donor-derived cells are sequentially followed.
Through day +100

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Event free survival (EFS)
Time Frame: At 2 years post transplant
-Death, disease recurrence or progression, and graft failure are considered events
At 2 years post transplant
Change in Lansky/Karnofsky performance score
Time Frame: Day +14, Day +30, Day +60, Day +100, Day +180, Day +365, +18 months, and +24 months
  • Lansky is used for participants <15.99 years of age
  • Karnofsky is used for participants >16.00 years of age
Day +14, Day +30, Day +60, Day +100, Day +180, Day +365, +18 months, and +24 months
Incidence and severity of acute GVHD
Time Frame: Weekly through day +100
-Incidence and severity of acute GVHD as graded according to the NIH consensus criteria. Severe aGVHD (Grades III-IV) is considered an event.
Weekly through day +100
Incidence and severity of chronic GVHD
Time Frame: Day 101 through 24 months
Incidence and severity of acute GVHD as graded according to the NIH consensus criteria. Severe cGHVD is considered an event.
Day 101 through 24 months
Number of neurologic toxicities
Time Frame: Through 24 months
-As evidence by clinical assessment and/or imaging at physician discretion
Through 24 months
Number of pulmonary toxicities
Time Frame: Through 24 months
-As determined by clinical context and serial assessment of TLC, FVC, FEV1, DLCO, or high-resolution CT Chest
Through 24 months
Number of cardiac toxicities
Time Frame: Through 24 months
-As determined by clinical assessment and EKG and/or echocardiogram
Through 24 months
Number of renal toxicities
Time Frame: Through 24 months
-As determined by clinical assessment and BUN, creatinine, urinalysis, estimated GFR, or renal scintigraphy
Through 24 months
Number of hepatic toxicities
Time Frame: Through 24 months
-As determined by clinical assessment and liver function tests
Through 24 months
Number of participants with infections
Time Frame: Through 24 months
Through 24 months
Number of metabolic toxicities
Time Frame: Through 24 months
-Metabolic toxicities will be collected using a complete metabolic panel
Through 24 months
Number of thyroid toxicities
Time Frame: Through 24 months
-Thyroid toxicities will be collected using a thyroid function test
Through 24 months
Immune reconstitution as measured by recovery of absolute neutrophil count
Time Frame: Over 24 months
Over 24 months
Immune reconstitution as measured by recovery of absolute monocyte count
Time Frame: Over 24 months
Over 24 months
Immune reconstitution as measured by regain of function of NK cell populations
Time Frame: Over 24 months
  • Immune reconstitution is defined as regain of function of donor-derived immunogenic cells. Immune reconstitution is to be measured by recovery of individual cellular compartments
  • Via flow cytometry
Over 24 months
Immune reconstitution as measured by regain of function of T cell populations
Time Frame: Through 24 months
  • Immune reconstitution is defined as regain of function of donor-derived immunogenic cells. Immune reconstitution is to be measured by recovery of individual cellular compartments
  • Via flow cytometry
Through 24 months
Immune reconstitution as measured by regain of function of B cell populations
Time Frame: Through 24 months
  • Immune reconstitution is defined as regain of function of donor-derived immunogenic cells. Immune reconstitution is to be measured by recovery of individual cellular compartments
  • Via flow cytometry
Through 24 months
Immune reconstitution as measured by regain of function of immunoglobulin G (IgG)
Time Frame: Over 24 months
  • Immune reconstitution is defined as regain of function of donor-derived immunogenic cells. Immune reconstitution is to be measured by recovery of individual cellular compartments
  • Via serum analysis
Over 24 months
Immune reconstitution as measured by regain of function of immunoglobulin A (IgA)
Time Frame: Over 24 months
  • Immune reconstitution is defined as regain of function of donor-derived immunogenic cells. Immune reconstitution is to be measured by recovery of individual cellular compartments
  • Via serum analysis
Over 24 months
Immune reconstitution as measured by regain of function of immunoglobulin M (IgM)
Time Frame: Over 24 months
  • Immune reconstitution is defined as regain of function of donor-derived immunogenic cells. Immune reconstitution is to be measured by recovery of individual cellular compartments
  • Via serum analysis
Over 24 months

Collaborators and Investigators

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

Investigators

  • Principal Investigator: Jeffrey Bednarski, M.D., Ph.D., Washington University School of Medicine

Publications and helpful links

The person responsible for entering information about the study voluntarily provides these publications. These may be about anything related to the study.

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)

November 3, 2022

Primary Completion (Estimated)

November 30, 2027

Study Completion (Estimated)

November 30, 2027

Study Registration Dates

First Submitted

August 11, 2021

First Submitted That Met QC Criteria

August 11, 2021

First Posted (Actual)

August 18, 2021

Study Record Updates

Last Update Posted (Actual)

March 12, 2024

Last Update Submitted That Met QC Criteria

March 8, 2024

Last Verified

March 1, 2024

More Information

Terms related to this study

Other Study ID Numbers

  • 202203051

Plan for Individual participant data (IPD)

Plan to Share Individual Participant Data (IPD)?

NO

Drug and device information, study documents

Studies a U.S. FDA-regulated drug product

No

Studies a U.S. FDA-regulated device product

Yes

product manufactured in and exported from the U.S.

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 Pediatric Hematologic Malignancies

Clinical Trials on Ex Vivo T-cell receptor alpha-beta and CD19+ Depletion using CliniMACs Plus

3
Subscribe