search
Back to results

Lentiviral Gene Therapy for X-linked Severe Combined Immunodeficiency

Primary Purpose

Severe Combined Immunodeficiency, X-Linked

Status
Recruiting
Phase
Phase 1
Locations
United Kingdom
Study Type
Interventional
Intervention
Lentiviral vector transduced CD34+ cells
Sponsored by
Great Ormond Street Hospital for Children NHS Foundation Trust
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional treatment trial for Severe Combined Immunodeficiency, X-Linked

Eligibility Criteria

8 Weeks - 5 Years (Child)MaleDoes not accept healthy volunteers

Inclusion Criteria:

  1. Diagnosis of SCID-X1 based on immunophenotype and lack of T cell function (proliferation to PHA <10% of the lower limit of normal for the laboratory) AND confirmed by a mutation in IL2RG
  2. Lack of an HLA identical (A, B, C, DR, DQ) related donor
  3. Age <5 years
  4. Signed informed consent
  5. Documentation of willingness to follow up for 15 years post-infusion
  6. If the patient has previously undergone allogeneic transplant or gene therapy, insufficiency of graft-derived T cell engraftment must be documented.
  7. Age at least 8 weeks of age by the time of busulfan administration

Exclusion Criteria:

  1. Patients with an active, therapy-resistant infection. Infections that are known to be highly morbid in SCID patients will be considered active and therapy-resistant if the infectious agent is repeatedly isolated despite a minimum of 2 weeks of appropriate therapy and is associated with significant organ dysfunction (including but not limited to abnormalities listed below).

    1. Mechanical ventilation including continuous positive airway pressure
    2. Abnormal liver function defined by AST and ALT >10 times the upper range of normal OR Bilirubin >2 mg/dL
    3. Shortening fraction on echocardiogram <25% or ejection fraction <50%
    4. Renal failure defined as glomerular filtration rate <30 ml/min/1.73 m2 or dialysis dependence
  2. Uncontrolled seizure disorder
  3. Encephalopathy
  4. Documented coexistence of any disorder known to affect DNA repair
  5. Diagnosis of active malignant disease other than EBV-associated lymphoproliferative disease
  6. Patients with evidence of infection with HIV-1
  7. Previous allogeneic transplant with cytoreductive chemotherapy
  8. Major (life-threatening) congenital anomalies. Examples of "major (life-threatening) congenital anomalies" include, but are not limited to: unrepaired cyanotic heart disease, hypoplastic lungs, anencephaly or other major central nervous system malformations, other severe non-repairable malformations of the gastrointestinal or genitourinary tracts that significantly impair organ function.
  9. Other conditions which in the opinion of the P.I. or Co-investigators, contra-indicate collection and/or infusion of transduced cells or indicate patient's inability to follow the protocol. These may include for example clinical ineligibility to receive anaesthesia, severe deterioration of clinical condition of the patient after collection of bone marrow but before infusion of transduced cells, or documented refusal or inability of the family to return for scheduled visits. There may be other unforeseen rare circumstances that would result in exclusion of the patient, such as sudden loss of legal guardianship.

Sites / Locations

  • Great Ormond Street Hospital for Children NHS Foundation TrustRecruiting

Arms of the Study

Arm 1

Arm Type

Experimental

Arm Label

Lentiviral vector transduced CD34+ cells

Arm Description

Single arm, non-randomised cohort of up to 5 patients with X-linked Severe Combined Immunodeficiency. CD34+ cells will be collected via bone marrow harvest or leukapheresis. The collected cells will then be purified, cultured and transduced with the G2SCID lentiviral vector. Transduced cells will be frozen. A minimum of 2.5 x 106/kg CD34+ cells after transduction with a minimum transduction efficiency of 0.7 copies/cell is required for infusion into the patient. The patient will receive non-myeloablative conditioning with intravenous busulfan the two or three days prior to cell infusion. The frozen cells will be thawed on the day of infusion and the cells administered according to hospital procedures. The patient will remain in hospital until sufficient cover of the patient's immune system

Outcomes

Primary Outcome Measures

Measure event-free survival after 1 year after gene transfer
Event-free survival at 1 year post-infusion. Events will include death, infusion of unmanipulated back-up product for failure of haematopoietic recovery, and allogeneic transplant performed for poor immune reconstitution
Measure T cell immune reconstitution: CD3+ T cell count
T cell reconstitution at 1 year post-infusion: CD3+ T cell count ≥300 cells/microliter in peripheral blood
Measure T cell immune reconstitution; gene marking
T cell reconstitution at 1 year post-infusion: Gene marking ≥0.1 copies/cell in sorted CD3+ T cells

Secondary Outcome Measures

Measure overall survival
Measure overall survival at 2 years post-infusion
Measure event-free survival
Measure event-free survival at 2 years post-infusion
Incidence of adverse events related to gene therapy
Incidence of adverse events related to gene therapy
Enumeration of absolute lymphocyte count determined by routine complete reconstitution
Enumeration of absolute lymphocyte count determined by routine complete blood counts (CBC)
Haematopoietic recovery after receipt of busulfan
Haematopoietic recovery is defined as absolute neutrophil count above 0.5 x10^9 /l for three consecutive days, achieved within 6 weeks following infusion.
Measure absolute numbers of T, B and NK lymphocytes
Absolute numbers of T, B and NK lymphocytes
Calculate percentage of naïve and memory T cell subsets
Percentage of naïve and memory T cell subsets
Measure laboratory results which correlates with efficacious immune reconstitution
Percentage of naïve and memory B cell subsets
Determine Freedom from immunoglobulin substitution for at least 9 months
Freedom from immunoglobulin substitution for at least 9 months
Measure serum immunoglobulin levels reconstitution
Serum immunoglobulin levels
Measure proliferation of lymphocytes to phytohaemagglutinin determined by titrated thymidine incorporation reconstitution
Proliferation of lymphocytes to phytohaemagglutinin determined by titrated thymidine incorporation
Measure antigen specific antibody titres to tetanus toxoid reconstitution
Measure antigen specific antibody titres to tetanus toxoid
Measure T cell receptor excision circles (TREC)
Measure T cell receptor excision circles (TREC)
Measure T cell receptor Vb family usage
Measure T cell receptor Vb family usage
To assess the efficacy of stem cell transduction/engraftment by measuring the frequency of gene marking in peripheral blood cells
Gene marking in specific lineages of peripheral blood cells. Genomic DNA isolated from each population will be assayed for VCN by quantitative PCR (qPCR). The results will be aggregated to determine the effectiveness of gene marking in the peripheral blood cells.
Measure clonal diversity of vector integrants
Clonal diversity will be quantitated and used to estimate the number of transduced haematopoietic stem cells that have engrafted in the subjects. Number of sequence reads and unique integration sites will be assessed to quantify population clone diversity, distribution of integration sites and relative abundance.

Full Information

First Posted
February 22, 2018
Last Updated
October 11, 2023
Sponsor
Great Ormond Street Hospital for Children NHS Foundation Trust
search

1. Study Identification

Unique Protocol Identification Number
NCT03601286
Brief Title
Lentiviral Gene Therapy for X-linked Severe Combined Immunodeficiency
Official Title
Phase I/II Study of Lentiviral Gene Transfer for SCID-X1 With Low Dose Targeted Busulfan
Study Type
Interventional

2. Study Status

Record Verification Date
October 2023
Overall Recruitment Status
Recruiting
Study Start Date
December 21, 2018 (Actual)
Primary Completion Date
August 2026 (Anticipated)
Study Completion Date
August 2026 (Anticipated)

3. Sponsor/Collaborators

Responsible Party, by Official Title
Sponsor
Name of the Sponsor
Great Ormond Street Hospital for Children NHS Foundation Trust

4. Oversight

Studies a U.S. FDA-regulated Drug Product
No
Studies a U.S. FDA-regulated Device Product
No
Data Monitoring Committee
Yes

5. Study Description

Brief Summary
Severe combined immunodeficiency disorder (SCID) is a heterogeneous group of inherited disorders characterized by a profound reduction or absence of T lymphocyte function, resulting in lack of both cellular and humoral immunity. SCID arises from a variety of molecular defects which affect lymphocyte development and function. The most common form of SCID is an X-linked form (SCID-X1), which accounts for 30-50% of all cases. SCID-X1 is caused by defects in the common cytokine receptor gamma chain, which was originally identified as a component of the high affinity interleukin-2 receptor (IL2RG). Allogeneic haematopoietic stem cell transplantation (HSCT), which replaces the patient's bone marrow with that of a healthy donor, is the only treatment that definitively restores the normal function of the bone marrow. HSCT is the first choice of treatment for patients with signs of bone marrow failure and a fully-matched related donor. However, patients without a fully-matched related donor have much worse overall outcomes from HSCT. This study will investigate whether patients with SCID-X1 without a fully matched related donor may benefit from gene therapy. To do this the investigators propose to perform a phase I/II clinical trial to evaluate the safety and efficacy (effect) of gene therapy for SCID-X1 patients using a lentivirus delivery system containing the IL2RG gene. Up to 5 eligible SCID-X1 patients will undergo mobilisation and harvest of their haematopoietic stem precursor cells (HPSCs). In the laboratory the disabled lentivirus will be used to insert a normal human IL2RG gene into the patient's harvested HPSCs. Patients will receive chemotherapy conditioning prior to cell infusion, in order to enhance grafting. The genetically corrected stem cells will then be re-infused into the patient. Patients will be followed up for 2 years. This trial will determine whether gene therapy for SCID-X1 using a lentiviral vector is safe, feasible and effective

6. Conditions and Keywords

Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
Severe Combined Immunodeficiency, X-Linked

7. Study Design

Primary Purpose
Treatment
Study Phase
Phase 1
Interventional Study Model
Single Group Assignment
Masking
None (Open Label)
Allocation
N/A
Enrollment
5 (Anticipated)

8. Arms, Groups, and Interventions

Arm Title
Lentiviral vector transduced CD34+ cells
Arm Type
Experimental
Arm Description
Single arm, non-randomised cohort of up to 5 patients with X-linked Severe Combined Immunodeficiency. CD34+ cells will be collected via bone marrow harvest or leukapheresis. The collected cells will then be purified, cultured and transduced with the G2SCID lentiviral vector. Transduced cells will be frozen. A minimum of 2.5 x 106/kg CD34+ cells after transduction with a minimum transduction efficiency of 0.7 copies/cell is required for infusion into the patient. The patient will receive non-myeloablative conditioning with intravenous busulfan the two or three days prior to cell infusion. The frozen cells will be thawed on the day of infusion and the cells administered according to hospital procedures. The patient will remain in hospital until sufficient cover of the patient's immune system
Intervention Type
Drug
Intervention Name(s)
Lentiviral vector transduced CD34+ cells
Other Intervention Name(s)
G2SCID lentiviral vector transduced CD34+ cells
Intervention Description
Gene therapy for X-linked Severe Combined Immunodeficiency will be performed by introduction a normal copy of the IL2RG gene into the blood forming stem cells (CD34+ cells) of the patient's bone marrow by using a type of gene delivery system (in this trial called a lentiviral vector). The gene corrected cells are then transplanted back into the patient.
Primary Outcome Measure Information:
Title
Measure event-free survival after 1 year after gene transfer
Description
Event-free survival at 1 year post-infusion. Events will include death, infusion of unmanipulated back-up product for failure of haematopoietic recovery, and allogeneic transplant performed for poor immune reconstitution
Time Frame
1 year
Title
Measure T cell immune reconstitution: CD3+ T cell count
Description
T cell reconstitution at 1 year post-infusion: CD3+ T cell count ≥300 cells/microliter in peripheral blood
Time Frame
1 year
Title
Measure T cell immune reconstitution; gene marking
Description
T cell reconstitution at 1 year post-infusion: Gene marking ≥0.1 copies/cell in sorted CD3+ T cells
Time Frame
1 year
Secondary Outcome Measure Information:
Title
Measure overall survival
Description
Measure overall survival at 2 years post-infusion
Time Frame
2 years
Title
Measure event-free survival
Description
Measure event-free survival at 2 years post-infusion
Time Frame
2 years
Title
Incidence of adverse events related to gene therapy
Description
Incidence of adverse events related to gene therapy
Time Frame
up to 2 years post-infusion of gene therapy
Title
Enumeration of absolute lymphocyte count determined by routine complete reconstitution
Description
Enumeration of absolute lymphocyte count determined by routine complete blood counts (CBC)
Time Frame
up to 2 years post-infusion of gene therapy
Title
Haematopoietic recovery after receipt of busulfan
Description
Haematopoietic recovery is defined as absolute neutrophil count above 0.5 x10^9 /l for three consecutive days, achieved within 6 weeks following infusion.
Time Frame
up to 6 weeks post-infusion of gene therapy
Title
Measure absolute numbers of T, B and NK lymphocytes
Description
Absolute numbers of T, B and NK lymphocytes
Time Frame
up to 2 years post-infusion of gene therapy
Title
Calculate percentage of naïve and memory T cell subsets
Description
Percentage of naïve and memory T cell subsets
Time Frame
up to 2 years post-infusion of gene therapy
Title
Measure laboratory results which correlates with efficacious immune reconstitution
Description
Percentage of naïve and memory B cell subsets
Time Frame
up to 2 years post-infusion of gene therapy
Title
Determine Freedom from immunoglobulin substitution for at least 9 months
Description
Freedom from immunoglobulin substitution for at least 9 months
Time Frame
2 years post-infusion of gene therapy
Title
Measure serum immunoglobulin levels reconstitution
Description
Serum immunoglobulin levels
Time Frame
up to 2 years post-infusion of gene therapy
Title
Measure proliferation of lymphocytes to phytohaemagglutinin determined by titrated thymidine incorporation reconstitution
Description
Proliferation of lymphocytes to phytohaemagglutinin determined by titrated thymidine incorporation
Time Frame
up to 2 years post-infusion of gene therapy
Title
Measure antigen specific antibody titres to tetanus toxoid reconstitution
Description
Measure antigen specific antibody titres to tetanus toxoid
Time Frame
up to 2 years post-infusion of gene therapy
Title
Measure T cell receptor excision circles (TREC)
Description
Measure T cell receptor excision circles (TREC)
Time Frame
up to 2 years post-infusion of gene therapy
Title
Measure T cell receptor Vb family usage
Description
Measure T cell receptor Vb family usage
Time Frame
up to 2 years post-infusion of gene therapy
Title
To assess the efficacy of stem cell transduction/engraftment by measuring the frequency of gene marking in peripheral blood cells
Description
Gene marking in specific lineages of peripheral blood cells. Genomic DNA isolated from each population will be assayed for VCN by quantitative PCR (qPCR). The results will be aggregated to determine the effectiveness of gene marking in the peripheral blood cells.
Time Frame
up to 2 years post-infusion of gene therapy
Title
Measure clonal diversity of vector integrants
Description
Clonal diversity will be quantitated and used to estimate the number of transduced haematopoietic stem cells that have engrafted in the subjects. Number of sequence reads and unique integration sites will be assessed to quantify population clone diversity, distribution of integration sites and relative abundance.
Time Frame
up to 2 years post-infusion of gene therapy
Other Pre-specified Outcome Measures:
Title
Correlation of potential biomarkers of humoral immune reconstitution with freedom from intravenous immunoglobulin substitution and antibody response to tetanus at 2 years post infusion including: Gene marking in B cells and B cell phenotype.
Description
Correlation of potential biomarkers of humoral immune reconstitution at 6 months, 1 year, 2 years post infusion with freedom from intravenous immunoglobulin substitution and antibody response to tetanus at 2 years post infusion including: Gene marking in B cells and B cell phenotype.
Time Frame
at 6 month, 12 month and 2 years post-infusion of gene therapy
Title
Correlation of busulfan area-under-the-curve measurements prior to infusion with freedom from intravenous immunoglobulin substitution and antibody response to tetanus at 2 years post-infusion and other markers of humoral immune reconstitution
Description
Correlation of busulfan area-under-the-curve measurements prior to infusion with freedom from intravenous immunoglobulin substitution and antibody response to tetanus at 2 years post-infusion and other markers of humoral immune reconstitution
Time Frame
2 years post-infusion of gene therapy
Title
Evidence of insertion site sharing between 2 or more lineages at 1 year and 2 years post infusion
Description
Evidence of insertion site sharing between 2 or more lineages at 1 year and 2 years post infusion
Time Frame
1 year and 2 years post infusion of gene therapy
Title
Correlation of gene marking and insertion site sharing in expanded peripheral blood CD34+ cells with peripheral blood mature cell samples at 1 year and 2 years post infusion
Description
Correlation of gene marking and insertion site sharing in expanded peripheral blood CD34+ cells with peripheral blood mature cell samples at 1 year and 2 years post infusion
Time Frame
1 year and 2 years post infusion of gene therapy
Title
Description of T cell receptor and B cell receptor repertoire before and after infusion
Description
Description of T cell receptor and B cell receptor repertoire before and after infusion
Time Frame
Pre-harvest, 3 month, 6 month, 12 month and 2 years post infusion of gene therapy
Title
Description of NK cell function and phenotype before and after infusion
Description
Description of NK cell function and phenotype before and after infusion
Time Frame
Pre-harvest, 3 month, 6 month, 12 month and 2 years post infusion of gene therapy

10. Eligibility

Sex
Male
Gender Based
Yes
Gender Eligibility Description
As SCID-X1 is an X-linked disorder, women/girls will not be enrolled.
Minimum Age & Unit of Time
8 Weeks
Maximum Age & Unit of Time
5 Years
Accepts Healthy Volunteers
No
Eligibility Criteria
Inclusion Criteria: Diagnosis of SCID-X1 based on immunophenotype and lack of T cell function (proliferation to PHA <10% of the lower limit of normal for the laboratory) AND confirmed by a mutation in IL2RG Lack of an HLA identical (A, B, C, DR, DQ) related donor Age <5 years Signed informed consent Documentation of willingness to follow up for 15 years post-infusion If the patient has previously undergone allogeneic transplant or gene therapy, insufficiency of graft-derived T cell engraftment must be documented. Age at least 8 weeks of age by the time of busulfan administration Exclusion Criteria: Patients with an active, therapy-resistant infection. Infections that are known to be highly morbid in SCID patients will be considered active and therapy-resistant if the infectious agent is repeatedly isolated despite a minimum of 2 weeks of appropriate therapy and is associated with significant organ dysfunction (including but not limited to abnormalities listed below). Mechanical ventilation including continuous positive airway pressure Abnormal liver function defined by AST and ALT >10 times the upper range of normal OR Bilirubin >2 mg/dL Shortening fraction on echocardiogram <25% or ejection fraction <50% Renal failure defined as glomerular filtration rate <30 ml/min/1.73 m2 or dialysis dependence Uncontrolled seizure disorder Encephalopathy Documented coexistence of any disorder known to affect DNA repair Diagnosis of active malignant disease other than EBV-associated lymphoproliferative disease Patients with evidence of infection with HIV-1 Previous allogeneic transplant with cytoreductive chemotherapy Major (life-threatening) congenital anomalies. Examples of "major (life-threatening) congenital anomalies" include, but are not limited to: unrepaired cyanotic heart disease, hypoplastic lungs, anencephaly or other major central nervous system malformations, other severe non-repairable malformations of the gastrointestinal or genitourinary tracts that significantly impair organ function. Other conditions which in the opinion of the P.I. or Co-investigators, contra-indicate collection and/or infusion of transduced cells or indicate patient's inability to follow the protocol. These may include for example clinical ineligibility to receive anaesthesia, severe deterioration of clinical condition of the patient after collection of bone marrow but before infusion of transduced cells, or documented refusal or inability of the family to return for scheduled visits. There may be other unforeseen rare circumstances that would result in exclusion of the patient, such as sudden loss of legal guardianship.
Central Contact Person:
First Name & Middle Initial & Last Name or Official Title & Degree
Claire Booth, Dr
Phone
0207 905 2198
Email
c.booth@ucl.ac.uk
First Name & Middle Initial & Last Name or Official Title & Degree
Karen Oprych, Dr
Email
k.gladwin@ucl.ac.uk
Overall Study Officials:
First Name & Middle Initial & Last Name & Degree
Claire Booth, Dr
Organizational Affiliation
UCL Great Ormond Street Institute of Child Health
Official's Role
Principal Investigator
First Name & Middle Initial & Last Name & Degree
Adrian Thrasher, Prof
Organizational Affiliation
UCL Great Ormond Street Institute of Child Health
Official's Role
Principal Investigator
Facility Information:
Facility Name
Great Ormond Street Hospital for Children NHS Foundation Trust
City
London
State/Province
Greater London
ZIP/Postal Code
WC1N 3JH
Country
United Kingdom
Individual Site Status
Recruiting
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Claire Booth, MBBS, MRCPCH, MSc, PhD
Email
c.booth@ucl.ac.uk
First Name & Middle Initial & Last Name & Degree
Karen Oprych, PhD
Email
k.gladwin@ucl.ac.uk
First Name & Middle Initial & Last Name & Degree
Claire Booth, MBBS, MRCPCH, MSc, PhD

12. IPD Sharing Statement

Plan to Share IPD
No

Learn more about this trial

Lentiviral Gene Therapy for X-linked Severe Combined Immunodeficiency

We'll reach out to this number within 24 hrs