Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Nat Commun ; 15(1): 974, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38321023

ABSTRACT

Invariant natural killer T (iNKT) cells, a unique T cell population, lend themselves for use as adoptive therapy due to diverse roles in orchestrating immune responses. Originally developed for use in cancer, agenT-797 is a donor-unrestricted allogeneic ex vivo expanded iNKT cell therapy. We conducted an open-label study in virally induced acute respiratory distress syndrome (ARDS) caused by the severe acute respiratory syndrome-2 virus (trial registration NCT04582201). Here we show that agenT-797 rescues exhausted T cells and rapidly activates both innate and adaptive immunity. In 21 ventilated patients including 5 individuals receiving veno-venous extracorporeal membrane oxygenation (VV-ECMO), there are no dose-limiting toxicities. We observe an anti-inflammatory systemic cytokine response and infused iNKT cells are persistent during follow-up, inducing only transient donor-specific antibodies. Clinical signals of associated survival and prevention of secondary infections are evident. Cellular therapy using off-the-shelf iNKT cells is safe, can be rapidly scaled and is associated with an anti-inflammatory response. The safety and therapeutic potential of iNKT cells across diseases including infections and cancer, warrants randomized-controlled trials.


Subject(s)
Natural Killer T-Cells , Neoplasms , Respiratory Distress Syndrome , Humans , Cytokines/metabolism , Anti-Inflammatory Agents
2.
Sci Adv ; 9(33): eadg6470, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37595035

ABSTRACT

To treat unilateral limbal stem cell (LSC) deficiency, we developed cultivated autologous limbal epithelial cells (CALEC) using an innovative xenobiotic-free, serum-free, antibiotic-free, two-step manufacturing process for LSC isolation and expansion onto human amniotic membrane with rigorous quality control in a good manufacturing practices facility. Limbal biopsies were used to generate CALEC constructs, and final grafts were evaluated by noninvasive scanning microscopy and tested for viability and sterility. Cultivated cells maintained epithelial cell phenotype with colony-forming and proliferative capacities. Analysis of LSC biomarkers showed preservation of "stemness." After preclinical development, a phase 1 clinical trial enrolled five patients with unilateral LSC deficiency. Four of these patients received CALEC transplants, establishing preliminary feasibility. Clinical case histories are reported, with no primary safety events. On the basis of these results, a second recruitment phase of the trial was opened to provide longer term safety and efficacy data on more patients.


Subject(s)
Anti-Bacterial Agents , Limbal Stem Cell Deficiency , Humans , Feasibility Studies , Biopsy , Commerce , Epithelial Cells
3.
Blood Adv ; 7(5): 768-777, 2023 03 14.
Article in English | MEDLINE | ID: mdl-35468618

ABSTRACT

Relapsed and refractory multiple myeloma (RRMM) is a plasma cell neoplasm defined by progressively refractory disease necessitating chronic and increasingly intensive therapy. Despite recent advances, limited treatment options exist for RRMM. This single-arm, open label phase 1 study aimed to evaluate the safety of novel B-cell maturation antigen (BCMA)-targeting chimeric antigen receptor (CAR) T construct that leverages a completely synthetic antigen-binding domain (CART-ddBCMA), which was specifically engineered to reduce immunogenicity and improve CAR cell surface stability. Thirteen patients ≥18 years with RRMM who received at least 3 prior regimens of systemic therapy were enrolled in the study. Patients received a single dose of 100 × 106 CART-ddBCMA (DL1) or 300 × 106 CART-ddBCMA (DL2) following standard lymphodepleting chemotherapy. The primary endpoints of the study were to evaluate the incidence of treatment emergent adverse events, including dose-limiting toxicities, and establish a recommended phase 2 dose. Results showed that CART-ddBCMA was well tolerated and demonstrated a favorable toxicity profile. Only 1 case of grade ≥3 cytokine release syndrome and 1 case of immune effector cell-associated neurotoxicity were reported; both were at DL2 and were manageable with standard treatment. No atypical neurological toxicities and Parkinson disease-like movement disorders were observed. The maximum tolerated dose was not reached. All infused patients responded to CART-ddBCMA, and 9/12 (75%) patients achieved complete response/stringent complete response. Responses deepened over time, and at the time of last data-cut (median follow-up 56 weeks), 8/9 (89%) evaluable patients achieved minimal residual disease negativity. In conclusion, the findings demonstrate the safety of CART-ddBCMA cells and document durable responses to CART-ddBCMA in patients with RRMM. This trial was registered at www.clinicaltrials.gov as #NCT04155749.


Subject(s)
Multiple Myeloma , Neurotoxicity Syndromes , Receptors, Chimeric Antigen , Humans , Multiple Myeloma/drug therapy , Lymphocytes , Receptors, Chimeric Antigen/therapeutic use
4.
Blood ; 138(15): 1304-1316, 2021 10 14.
Article in English | MEDLINE | ID: mdl-33974038

ABSTRACT

Patients lacking functional adenosine deaminase activity have severe combined immunodeficiency (ADA SCID), which can be treated with ADA enzyme replacement therapy (ERT), allogeneic hematopoietic stem cell transplantation (HSCT), or autologous HSCT with gene-corrected cells (gene therapy [GT]). A cohort of 10 ADA SCID patients, aged 3 months to 15 years, underwent GT in a phase 2 clinical trial between 2009 and 2012. Autologous bone marrow CD34+ cells were transduced ex vivo with the MND (myeloproliferative sarcoma virus, negative control region deleted, dl587rev primer binding site)-ADA gammaretroviral vector (gRV) and infused following busulfan reduced-intensity conditioning. These patients were monitored in a long-term follow-up protocol over 8 to 11 years. Nine of 10 patients have sufficient immune reconstitution to protect against serious infections and have not needed to resume ERT or proceed to secondary allogeneic HSCT. ERT was restarted 6 months after GT in the oldest patient who had no evidence of benefit from GT. Four of 9 evaluable patients with the highest gene marking and B-cell numbers remain off immunoglobulin replacement therapy and responded to vaccines. There were broad ranges of responses in normalization of ADA enzyme activity and adenine metabolites in blood cells and levels of cellular and humoral immune reconstitution. Outcomes were generally better in younger patients and those receiving higher doses of gene-marked CD34+ cells. No patient experienced a leukoproliferative event after GT, despite persisting prominent clones with vector integrations adjacent to proto-oncogenes. These long-term findings demonstrate enduring efficacy of GT for ADA SCID but also highlight risks of genotoxicity with gRVs. This trial was registered at www.clinicaltrials.gov as #NCT00794508.


Subject(s)
Agammaglobulinemia/therapy , Genetic Therapy , Severe Combined Immunodeficiency/therapy , Adenosine Deaminase/genetics , Adolescent , Agammaglobulinemia/genetics , Child , Child, Preschool , Follow-Up Studies , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation/methods , Humans , Infant , Severe Combined Immunodeficiency/genetics , Transplantation, Autologous/methods , Treatment Outcome
5.
N Engl J Med ; 384(21): 2002-2013, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33974366

ABSTRACT

BACKGROUND: Severe combined immunodeficiency due to adenosine deaminase (ADA) deficiency (ADA-SCID) is a rare and life-threatening primary immunodeficiency. METHODS: We treated 50 patients with ADA-SCID (30 in the United States and 20 in the United Kingdom) with an investigational gene therapy composed of autologous CD34+ hematopoietic stem and progenitor cells (HSPCs) transduced ex vivo with a self-inactivating lentiviral vector encoding human ADA. Data from the two U.S. studies (in which fresh and cryopreserved formulations were used) at 24 months of follow-up were analyzed alongside data from the U.K. study (in which a fresh formulation was used) at 36 months of follow-up. RESULTS: Overall survival was 100% in all studies up to 24 and 36 months. Event-free survival (in the absence of reinitiation of enzyme-replacement therapy or rescue allogeneic hematopoietic stem-cell transplantation) was 97% (U.S. studies) and 100% (U.K. study) at 12 months; 97% and 95%, respectively, at 24 months; and 95% (U.K. study) at 36 months. Engraftment of genetically modified HSPCs persisted in 29 of 30 patients in the U.S. studies and in 19 of 20 patients in the U.K. study. Patients had sustained metabolic detoxification and normalization of ADA activity levels. Immune reconstitution was robust, with 90% of the patients in the U.S. studies and 100% of those in the U.K. study discontinuing immunoglobulin-replacement therapy by 24 months and 36 months, respectively. No evidence of monoclonal expansion, leukoproliferative complications, or emergence of replication-competent lentivirus was noted, and no events of autoimmunity or graft-versus-host disease occurred. Most adverse events were of low grade. CONCLUSIONS: Treatment of ADA-SCID with ex vivo lentiviral HSPC gene therapy resulted in high overall and event-free survival with sustained ADA expression, metabolic correction, and functional immune reconstitution. (Funded by the National Institutes of Health and others; ClinicalTrials.gov numbers, NCT01852071, NCT02999984, and NCT01380990.).


Subject(s)
Agammaglobulinemia/therapy , Genetic Therapy/methods , Genetic Vectors , Hematopoietic Stem Cell Transplantation , Lentivirus/genetics , Severe Combined Immunodeficiency/therapy , Adenosine Deaminase/deficiency , Adolescent , Child , Child, Preschool , Genetic Therapy/adverse effects , Humans , Infant , Lymphocyte Count , Progression-Free Survival , Prospective Studies , Transplantation, Autologous
6.
N Engl J Med ; 384(3): 205-215, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33283990

ABSTRACT

BACKGROUND: Sickle cell disease is characterized by hemolytic anemia, pain, and progressive organ damage. A high level of erythrocyte fetal hemoglobin (HbF) comprising α- and γ-globins may ameliorate these manifestations by mitigating sickle hemoglobin polymerization and erythrocyte sickling. BCL11A is a repressor of γ-globin expression and HbF production in adult erythrocytes. Its down-regulation is a promising therapeutic strategy for induction of HbF. METHODS: We enrolled patients with sickle cell disease in a single-center, open-label pilot study. The investigational therapy involved infusion of autologous CD34+ cells transduced with the BCH-BB694 lentiviral vector, which encodes a short hairpin RNA (shRNA) targeting BCL11A mRNA embedded in a microRNA (shmiR), allowing erythroid lineage-specific knockdown. Patients were assessed for primary end points of engraftment and safety and for hematologic and clinical responses to treatment. RESULTS: As of October 2020, six patients had been followed for at least 6 months after receiving BCH-BB694 gene therapy; median follow-up was 18 months (range, 7 to 29). All patients had engraftment, and adverse events were consistent with effects of the preparative chemotherapy. All the patients who could be fully evaluated achieved robust and stable HbF induction (percentage HbF/(F+S) at most recent follow-up, 20.4 to 41.3%), with HbF broadly distributed in red cells (F-cells 58.9 to 93.6% of untransfused red cells) and HbF per F-cell of 9.0 to 18.6 pg per cell. Clinical manifestations of sickle cell disease were reduced or absent during the follow-up period. CONCLUSIONS: This study validates BCL11A inhibition as an effective target for HbF induction and provides preliminary evidence that shmiR-based gene knockdown offers a favorable risk-benefit profile in sickle cell disease. (Funded by the National Institutes of Health; ClinicalTrials.gov number, NCT03282656).


Subject(s)
Anemia, Sickle Cell/therapy , Fetal Hemoglobin/biosynthesis , Genetic Therapy , RNA Interference , Repressor Proteins/genetics , gamma-Globins/metabolism , Adolescent , Adult , Anemia, Sickle Cell/genetics , Child , Down-Regulation , Female , Fetal Hemoglobin/genetics , Gene Knockdown Techniques , Genetic Vectors , Humans , Male , Pilot Projects , RNA, Small Interfering , Repressor Proteins/metabolism , Transplantation, Autologous , Young Adult , gamma-Globins/genetics
7.
Biol Blood Marrow Transplant ; 26(10): 1819-1827, 2020 10.
Article in English | MEDLINE | ID: mdl-32653625

ABSTRACT

The pharmacokinetics of low-dose busulfan (BU) were investigated as a nonmyeloablative conditioning regimen for autologous gene therapy (GT) in pediatric subjects with adenosine deaminase-deficient severe combined immunodeficiency disease (ADA SCID). In 3 successive clinical trials, which included either γ-retroviral (γ-RV) or lentiviral (LV) vectors, subjects were conditioned with BU using different dosing nomograms. The first cohort received BU doses based on body surface area (BSA), the second cohort received doses based on actual body weight (ABW), and in the third cohort, therapeutic drug monitoring (TDM) was used to target a specific area under the concentration-time curve (AUC). Neither BSA-based nor ABW-based dosing achieved a consistent cumulative BU AUC; in contrast, TDM-based dosing led to more consistent AUC. BU clearance increased as subject age increased from birth to 18 months. However, weight and age alone were insufficient to accurately predict the dose that would consistently achieve a target AUC. Furthermore, various clinical, laboratory, and genetic factors (eg, genotypes for glutathione-S-transferase isozymes known to participate in BU metabolism) were analyzed, but no single finding predicted subjects with rapid versus slow clearance. Analysis of BU AUC and the postengraftment vector copy number (VCN) in granulocytes, a surrogate marker of the level of engrafted gene-modified hematopoietic stem and progenitor cells (HSPCs), demonstrated gene marking at levels sufficient for therapeutic benefit in the subjects who had achieved the target BU AUC. Although many factors determine the ultimate engraftment following GT, this work demonstrates that the BU AUC correlated with the eventual level of engrafted gene-modified HSPCs within a vector group (γ-RV versus LV), with significantly higher levels of granulocyte VCN in the recipients of LV-modified grafts compared to recipients of γ-RV-transduced grafts. Taken together, these findings provide insight into low-dose BU pharmacokinetics in the unique setting of autologous GT for ADA SCID, and these dosing principles may be applied to future GT trials using low-dose BU to open the bone marrow niche.


Subject(s)
Hematopoietic Stem Cell Transplantation , Severe Combined Immunodeficiency , Adenosine Deaminase/genetics , Agammaglobulinemia , Busulfan , Child , Genetic Therapy , Humans , Infant , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy , Transplantation Conditioning
8.
Nat Med ; 26(2): 200-206, 2020 02.
Article in English | MEDLINE | ID: mdl-31988463

ABSTRACT

Chronic granulomatous disease (CGD) is a rare inherited disorder of phagocytic cells1,2. We report the initial results of nine severely affected X-linked CGD (X-CGD) patients who received ex vivo autologous CD34+ hematopoietic stem and progenitor cell-based lentiviral gene therapy following myeloablative conditioning in first-in-human studies (trial registry nos. NCT02234934 and NCT01855685). The primary objectives were to assess the safety and evaluate the efficacy and stability of biochemical and functional reconstitution in the progeny of engrafted cells at 12 months. The secondary objectives included the evaluation of augmented immunity against bacterial and fungal infection, as well as assessment of hematopoietic stem cell transduction and engraftment. Two enrolled patients died within 3 months of treatment from pre-existing comorbidities. At 12 months, six of the seven surviving patients demonstrated stable vector copy numbers (0.4-1.8 copies per neutrophil) and the persistence of 16-46% oxidase-positive neutrophils. There was no molecular evidence of either clonal dysregulation or transgene silencing. Surviving patients have had no new CGD-related infections, and six have been able to discontinue CGD-related antibiotic prophylaxis. The primary objective was met in six of the nine patients at 12 months follow-up, suggesting that autologous gene therapy is a promising approach for CGD patients.


Subject(s)
Chromosomes, Human, X , Genetic Therapy/methods , Granulomatous Disease, Chronic/genetics , Lentivirus/genetics , Adolescent , Antigens, CD34/genetics , Child , Child, Preschool , Comorbidity , Gene Silencing , Genes, Regulator , Genetic Vectors , Granulomatous Disease, Chronic/therapy , Hematopoietic Stem Cells/cytology , Humans , Male , NADPH Oxidases/genetics , Neutrophils/metabolism , Patient Safety , Promoter Regions, Genetic , Transplantation Conditioning , Treatment Outcome , United Kingdom , United States , Young Adult
9.
J Clin Invest ; 127(5): 1689-1699, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28346229

ABSTRACT

BACKGROUND: Autologous hematopoietic stem cell transplantation (HSCT) of gene-modified cells is an alternative to enzyme replacement therapy (ERT) and allogeneic HSCT that has shown clinical benefit for adenosine deaminase-deficient (ADA-deficient) SCID when combined with reduced intensity conditioning (RIC) and ERT cessation. Clinical safety and therapeutic efficacy were evaluated in a phase II study. METHODS: Ten subjects with confirmed ADA-deficient SCID and no available matched sibling or family donor were enrolled between 2009 and 2012 and received transplantation with autologous hematopoietic CD34+ cells that were modified with the human ADA cDNA (MND-ADA) γ-retroviral vector after conditioning with busulfan (90 mg/m2) and ERT cessation. Subjects were followed from 33 to 84 months at the time of data analysis. Safety of the procedure was assessed by recording the number of adverse events. Efficacy was assessed by measuring engraftment of gene-modified hematopoietic stem/progenitor cells, ADA gene expression, and immune reconstitution. RESULTS: With the exception of the oldest subject (15 years old at enrollment), all subjects remained off ERT with normalized peripheral blood mononuclear cell (PBMC) ADA activity, improved lymphocyte numbers, and normal proliferative responses to mitogens. Three of nine subjects were able to discontinue intravenous immunoglobulin replacement therapy. The MND-ADA vector was persistently detected in PBMCs (vector copy number [VCN] = 0.1-2.6) and granulocytes (VCN = 0.01-0.3) through the most recent visits at the time of this writing. No patient has developed a leukoproliferative disorder or other vector-related clinical complication since transplant. CONCLUSION: These results demonstrate clinical therapeutic efficacy from gene therapy for ADA-deficient SCID, with an excellent clinical safety profile. TRIAL REGISTRATION: ClinicalTrials.gov NCT00794508. FUNDING: Food and Drug Administration Office of Orphan Product Development award, RO1 FD003005; NHLBI awards, PO1 HL73104 and Z01 HG000122; UCLA Clinical and Translational Science Institute awards, UL1RR033176 and UL1TR000124.


Subject(s)
Adenosine Deaminase/deficiency , Agammaglobulinemia , Gene Expression Regulation, Enzymologic , Genetic Therapy , Hematopoietic Stem Cell Transplantation , Severe Combined Immunodeficiency , Transduction, Genetic , Adenosine Deaminase/biosynthesis , Adenosine Deaminase/genetics , Adolescent , Agammaglobulinemia/enzymology , Agammaglobulinemia/genetics , Agammaglobulinemia/therapy , Autografts , Child , Child, Preschool , Female , Genetic Vectors , Humans , Infant , Male , Retroviridae , Severe Combined Immunodeficiency/enzymology , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy
10.
Blood ; 129(19): 2624-2635, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28351939

ABSTRACT

Retroviral gene therapy has proved efficacious for multiple genetic diseases of the hematopoietic system, but roughly half of clinical gene therapy trial protocols using gammaretroviral vectors have reported leukemias in some of the patients treated. In dramatic contrast, 39 adenosine deaminase-deficient severe combined immunodeficiency (ADA-SCID) patients have been treated with 4 distinct gammaretroviral vectors without oncogenic consequence. We investigated clonal dynamics and diversity in a cohort of 15 ADA-SCID children treated with gammaretroviral vectors and found clear evidence of genotoxicity, indicated by numerous common integration sites near proto-oncogenes and by increased abundance of clones with integrations near MECOM and LMO2 These clones showed stable behavior over multiple years and never expanded to the point of dominance or dysplasia. One patient developed a benign clonal dominance that could not be attributed to insertional mutagenesis and instead likely resulted from expansion of a transduced natural killer clone in response to chronic Epstein-Barr virus viremia. Clonal diversity and T-cell repertoire, measured by vector integration site sequencing and T-cell receptor ß-chain rearrangement sequencing, correlated significantly with the amount of busulfan preconditioning delivered to patients and to CD34+ cell dose. These data, in combination with results of other ADA-SCID gene therapy trials, suggest that disease background may be a crucial factor in leukemogenic potential of retroviral gene therapy and underscore the importance of cytoreductive conditioning in this type of gene therapy approach.


Subject(s)
Adenosine Deaminase/deficiency , Agammaglobulinemia/genetics , Agammaglobulinemia/therapy , Antineoplastic Agents, Alkylating/therapeutic use , Busulfan/therapeutic use , Gammaretrovirus/genetics , Genetic Therapy/methods , Genetic Vectors/therapeutic use , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy , Adaptor Proteins, Signal Transducing/genetics , Adenosine Deaminase/genetics , Agammaglobulinemia/pathology , Child , DNA-Binding Proteins/genetics , Genetic Vectors/genetics , Humans , LIM Domain Proteins/genetics , MDS1 and EVI1 Complex Locus Protein , Proto-Oncogene Proteins/genetics , Proto-Oncogenes/genetics , Severe Combined Immunodeficiency/pathology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Transcription Factors/genetics
11.
N Engl J Med ; 371(15): 1407-17, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25295500

ABSTRACT

BACKGROUND: In previous clinical trials involving children with X-linked severe combined immunodeficiency (SCID-X1), a Moloney murine leukemia virus-based γ-retrovirus vector expressing interleukin-2 receptor γ-chain (γc) complementary DNA successfully restored immunity in most patients but resulted in vector-induced leukemia through enhancer-mediated mutagenesis in 25% of patients. We assessed the efficacy and safety of a self-inactivating retrovirus for the treatment of SCID-X1. METHODS: We enrolled nine boys with SCID-X1 in parallel trials in Europe and the United States to evaluate treatment with a self-inactivating (SIN) γ-retrovirus vector containing deletions in viral enhancer sequences expressing γc (SIN-γc). RESULTS: All patients received bone marrow-derived CD34+ cells transduced with the SIN-γc vector, without preparative conditioning. After 12.1 to 38.7 months of follow-up, eight of the nine children were still alive. One patient died from an overwhelming adenoviral infection before reconstitution with genetically modified T cells. Of the remaining eight patients, seven had recovery of peripheral-blood T cells that were functional and led to resolution of infections. The patients remained healthy thereafter. The kinetics of CD3+ T-cell recovery was not significantly different from that observed in previous trials. Assessment of insertion sites in peripheral blood from patients in the current trial as compared with those in previous trials revealed significantly less clustering of insertion sites within LMO2, MECOM, and other lymphoid proto-oncogenes in our patients. CONCLUSIONS: This modified γ-retrovirus vector was found to retain efficacy in the treatment of SCID-X1. The long-term effect of this therapy on leukemogenesis remains unknown. (Funded by the National Institutes of Health and others; ClinicalTrials.gov numbers, NCT01410019, NCT01175239, and NCT01129544.).


Subject(s)
Gammaretrovirus/genetics , Genetic Therapy , Genetic Vectors , X-Linked Combined Immunodeficiency Diseases/therapy , Animals , Antigens, CD34 , DNA, Complementary/therapeutic use , Gene Expression , Gene Silencing , Genetic Therapy/adverse effects , Humans , Infant , Interleukin Receptor Common gamma Subunit/genetics , Male , Mice , Mutation , T-Lymphocytes/immunology , Transduction, Genetic , Transgenes/physiology , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/immunology
12.
Blood ; 120(18): 3635-46, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22968453

ABSTRACT

We conducted a gene therapy trial in 10 patients with adenosine deaminase (ADA)-deficient severe combined immunodeficiency using 2 slightly different retroviral vectors for the transduction of patients' bone marrow CD34(+) cells. Four subjects were treated without pretransplantation cytoreduction and remained on ADA enzyme-replacement therapy (ERT) throughout the procedure. Only transient (months), low-level (< 0.01%) gene marking was observed in PBMCs of 2 older subjects (15 and 20 years of age), whereas some gene marking of PBMC has persisted for the past 9 years in 2 younger subjects (4 and 6 years). Six additional subjects were treated using the same gene transfer protocol, but after withdrawal of ERT and administration of low-dose busulfan (65-90 mg/m(2)). Three of these remain well, off ERT (5, 4, and 3 years postprocedure), with gene marking in PBMC of 1%-10%, and ADA enzyme expression in PBMC near or in the normal range. Two subjects were restarted on ERT because of poor gene marking and immune recovery, and one had a subsequent allogeneic hematopoietic stem cell transplantation. These studies directly demonstrate the importance of providing nonmyeloablative pretransplantation conditioning to achieve therapeutic benefits with gene therapy for ADA-deficient severe combined immunodeficiency.


Subject(s)
Agammaglobulinemia/therapy , Bone Marrow Transplantation/methods , Genetic Therapy/methods , Genetic Vectors , Hematopoietic Stem Cell Transplantation/methods , Severe Combined Immunodeficiency/therapy , Adenosine Deaminase/deficiency , Adolescent , Antigens, CD34/metabolism , Child , Child, Preschool , Female , Humans , Infant , Male , Retroviridae/genetics , Transduction, Genetic , Transplantation Conditioning , Young Adult
13.
Sci Transl Med ; 3(97): 97ps36, 2011 Aug 24.
Article in English | MEDLINE | ID: mdl-21865536

ABSTRACT

Hematopoietic stem cell (HSC) transplantation may be curative for severe combined immunodeficiency (SCID). However, for a majority of infants with SCID a suitable donor is not available, and even with a matched donor, allogeneic HSC transplantation itself carries potential complications such as graft-versus-host disease as well as side effects from myelosuppressive chemotherapy. In the past decade, substantial advances have been made in the transplantation of gene-modified autologous HSCs, especially for two forms of SCID: X-linked SCID (SCID-X1) and adenosine deaminase (ADA)-deficient SCID. Two new reports in this issue of Science Translational Medicine add to the accumulating findings from gene therapy trials in Italy, France, and the United States that show clinical benefits of this alternative treatment.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Severe Combined Immunodeficiency/therapy , Adenosine Deaminase/deficiency , Agammaglobulinemia/therapy , Clinical Trials as Topic , France , Humans , Infant , Italy , United States , X-Linked Combined Immunodeficiency Diseases/therapy
14.
Blood ; 118(10): 2688-94, 2011 Sep 08.
Article in English | MEDLINE | ID: mdl-21725047

ABSTRACT

Genetic deficiency of adenosine deaminase (ADA) can cause profound lymphopenia and result in the clinical presentation of severe combined immune deficiency (SCID). However, because of the ubiquitous expression of ADA, ADA-deficient patients often present also with nonimmunologic clinical problems, affecting the skeletal, central nervous, endocrine, and gastrointestinal systems. We now report that myeloid dysplasia features and bone marrow hypocellularity are often found in patients with ADA-SCID. As a clinical correlate to this finding, we have observed vulnerability to antibiotic-induced myelotoxicity and prolonged neutropenia after nonmyeloablative chemotherapy. We have also noted that, in the absence of enzyme replacement therapy, absolute neutrophil counts of patients with ADA deficiency vary inversely with the accumulation of deoxynucleotides. These data have significant implications for the application of standard and investigational therapies to patients with ADA-SCID and support further studies to investigate the possibility that ADA deficiency is associated with a stem cell defect. These trials were registered at www.clinicaltrials.gov as #NCT00018018 and #NCT00006319.


Subject(s)
Adenosine Deaminase/deficiency , Agammaglobulinemia/complications , Bone Marrow/pathology , Myelodysplastic Syndromes/etiology , Severe Combined Immunodeficiency/complications , Adenosine Deaminase/genetics , Adolescent , Adult , Agammaglobulinemia/therapy , Bone Marrow Transplantation , Child , Child, Preschool , Female , Genetic Therapy , Humans , Infant , Male , Myelodysplastic Syndromes/therapy , Severe Combined Immunodeficiency/therapy , Young Adult
15.
Mol Ther ; 18(7): 1389-96, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20389286

ABSTRACT

Although pancreatic beta-cell transplantation may serve as a potential cure for diabetes mellitus (DM), limited donor tissue availability poses a major challenge. Thus, there is a great demand to find new sources for pancreatic beta-cells. Here, we present a lentiviral vector-based approach to achieve beta-cell proliferation through the beta-cell-specific activation of the hepatocyte growth factor (HGF)/cmet signaling pathway. The methodology is based on the beta-cell-specific expression of a ligand-inducible, chimeric receptor (F36Vcmet), under transcriptional control of the promoter from the human insulin gene, and its ability to induce HGF/cmet signaling in the presence of a synthetic ligand (AP20187). High transduction efficiency of human pancreatic islets was achieved utilizing this approach with chimeric receptor expression confined to the beta-cell population. In addition, specific proliferation of human pancreatic beta-cells was induced utilizing this approach. Selective, regulated beta-cell expansion may help to provide greater availability of cells for transplantation in patients with DM.


Subject(s)
Insulin-Secreting Cells/cytology , Adult , Animals , Cell Line, Tumor , Cell Proliferation , Genetic Vectors/genetics , HT29 Cells , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Insulin-Secreting Cells/metabolism , Lentivirus/genetics , Mice , Promoter Regions, Genetic/genetics , Tissue Culture Techniques
16.
Stem Cells Dev ; 15(1): 109-17, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16522168

ABSTRACT

Genetic modification of human embryonic stem cells (hESCs) is an important tool for understanding and influencing their biologic properties. At the present time, lentiviral vectors pseudotyped with the vesicular stomatitis virus G protein (VSV-G) have been most effective for stable gene transfer to hESCs. However, they also efficiently transduce murine embryonic fibroblasts (MEF), used to support the undifferentiated state of many commonly used hESC lines. Transduction of both the MEF as well as hESCs complicates analyses of gene transfer and expression. We made lentiviral vectors pseudotyped with envelope glycoproteins from retroviruses that have been shown to have more restricted transduction ranges and evaluated their specificity. Lentiviral vectors pseudotyped by the envelopes from either the gibbon ape leukemia virus (GALV) or the RD114 feline endogenous virus (RD114) specifically transduced hESCs to similar extents as VSV-G pseudotyped vectors, but did not transduce MEF. In addition, gene modfication by these pseudotyped lentiviral vectors was stably maintained throughout differentiation of hESCs in vitro. These pseudotyped lentiviral vectors may be valuable tools for efficient, specific and stable gene modification of hESCs.


Subject(s)
Cell Differentiation/genetics , Gene Transfer Techniques , Genetic Vectors , Hematopoietic Stem Cells/metabolism , Lentivirus/genetics , Amino Acid Transport System ASC/genetics , Animals , Cell Line , Glycoproteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Minor Histocompatibility Antigens
17.
J Virol ; 77(4): 2338-48, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12551971

ABSTRACT

Foamy viruses (FVs) are classified in the family Retroviridae, but recent data have shown that they are not conventional retroviruses. Notably, several characteristics of their particle replication strategies are more similar to those of hepatitis B virus (HBV) than those of typical retroviruses. Compared to conventional retroviruses, which require only Gag proteins for budding and release of virus-like particles (VLPs), both FV and HBV require Env proteins. In the case of HBV, Env (S protein) alone is sufficient to form subviral particles (SVPs). Because FVs also depend on Env for budding, we tested whether FV Env alone could produce SVPs. The Env proteins of FV and murine leukemia virus (MuLV) were both released into cell culture supernatants and migrated into isopycnic gradients; however, unlike MuLV Env, FV Env displayed characteristics of SVPs. FV Env particles were of greater density than those of MuLV (1.11 versus 1.07 g/ml, respectively), which strongly suggested that the released proteins of FV Env were particulate. When we examined FV SVPs by immunoelectron microscopy, we found particles that were consistent in morphology, size, and staining with gold beads, similar to FV VLPs and unlike the particle-like structures of MuLV Env, which were more consistent with vesicles produced from nonspecific membrane "blebbing." Taken together, our results demonstrated that FV Env alone is sufficient for particle budding. This finding is unique among retroviruses and further demonstrated the similarities between FV and HBV.


Subject(s)
Spumavirus/physiology , Viral Envelope Proteins/metabolism , Virion/physiology , Animals , Cell Line , Centrifugation, Density Gradient , Cricetinae , Culture Media , Humans , Microscopy, Electron , Spumavirus/metabolism , Viral Envelope Proteins/genetics , Virion/metabolism , Virus Assembly , Virus Shedding
SELECTION OF CITATIONS
SEARCH DETAIL
...