Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Denise A. Carbonaro is active.

Publication


Featured researches published by Denise A. Carbonaro.


Nature Medicine | 2003

Clonality analysis after retroviral-mediated gene transfer to CD34+ cells from the cord blood of ADA-deficient SCID neonates

Manfred Schmidt; Denise A. Carbonaro; Carsten Speckmann; Manuela Wissler; John F. Bohnsack; Melissa Elders; Bruce J. Aronow; Jan A. Nolta; Donald B. Kohn; Christof von Kalle

A clinical trial of retroviral-mediated transfer of the adenosine deaminase (ADA) gene into umbilical cord blood CD34+ cells was started in 1993. ADA-containing peripheral blood mononuclear cells (PBMCs) have persisted in patients from this trial, with T lymphocytes showing the highest prevalence of gene marking. To gain a greater understanding of the nature and number of the transduced cells that were engrafted, we used linear amplification–mediated PCR (LAM-PCR) to identify clonal vector proviral integrants. In one patient, a single vector integrant was predominant in T lymphocytes at a stable level over most of the eight-year time span analyzed and was also detected in some myeloid samples. T-cell clones with the predominant integrant, isolated after eight years, showed multiple patterns of T-cell receptor (TCR) gene rearrangement, indicating that a single pre-thymic stem or progenitor cell served as the source of the majority of the gene-marked cells over an extended period of time. It is important to distinguish the stable pattern of monoclonal gene marking that we observed here from the progressive increase of a T-cell clone with monoclonal gene marking that results from leukemic transformation, as observed in two subjects in a clinical trial of gene therapy for X-linked severe combined immunodeficiency (SCID).


Molecular Therapy | 2014

Preclinical Demonstration of Lentiviral Vector-mediated Correction of Immunological and Metabolic Abnormalities in Models of Adenosine Deaminase Deficiency

Denise A. Carbonaro; Lin Zhang; Xiangyang Jin; Claudia Montiel-Equihua; Sabine Geiger; Marlene Carmo; Aaron R. Cooper; Lynette Fairbanks; Michael L. Kaufman; Nj Sebire; Roger P. Hollis; Michael P. Blundell; Shantha Senadheera; Pei Yu Fu; Arineh Sahaghian; Rebecca Chan; Xiaoyan Wang; Kenneth Cornetta; Adrian J. Thrasher; Donald B. Kohn; H. Bobby Gaspar

Gene transfer into autologous hematopoietic stem cells by γ-retroviral vectors (gRV) is an effective treatment for adenosine deaminase (ADA)-deficient severe combined immunodeficiency (SCID). However, current gRV have significant potential for insertional mutagenesis as reported in clinical trials for other primary immunodeficiencies. To improve the efficacy and safety of ADA-SCID gene therapy (GT), we generated a self-inactivating lentiviral vector (LV) with a codon-optimized human cADA gene under the control of the short form elongation factor-1α promoter (LV EFS ADA). In ADA(-/-) mice, LV EFS ADA displayed high-efficiency gene transfer and sufficient ADA expression to rescue ADA(-/-) mice from their lethal phenotype with good thymic and peripheral T- and B-cell reconstitution. Human ADA-deficient CD34(+) cells transduced with 1-5 × 10(7) TU/ml had 1-3 vector copies/cell and expressed 1-2x of normal endogenous levels of ADA, as assayed in vitro and by transplantation into immune-deficient mice. Importantly, in vitro immortalization assays demonstrated that LV EFS ADA had significantly less transformation potential compared to gRV vectors, and vector integration-site analysis by nrLAM-PCR of transduced human cells grown in immune-deficient mice showed no evidence of clonal skewing. These data demonstrated that the LV EFS ADA vector can effectively transfer the human ADA cDNA and promote immune and metabolic recovery, while reducing the potential for vector-mediated insertional mutagenesis.


Blood | 2012

Gene therapy/bone marrow transplantation in ADA-deficient mice: Roles of enzyme-replacement therapy and cytoreduction

Denise A. Carbonaro; Xiangyang Jin; Xingchao Wang; Xiao-Jin Yu; Rozengurt N; Michael L. Kaufman; David W. Gjertson; Yang Zhou; Michael R. Blackburn; Donald B. Kohn

Gene therapy (GT) for adenosine deaminase-deficient severe combined immune deficiency (ADA-SCID) can provide significant long-term benefit when patients are given nonmyeloablative conditioning and ADA enzyme-replacement therapy (ERT) is withheld before autologous transplantation of γ-retroviral vector-transduced BM CD34+ cells. To determine the contributions of conditioning and discontinuation of ERT to the therapeutic effects, we analyzed these factors in Ada gene knockout mice (Ada(-/-)). Mice were transplanted with ADA-deficient marrow transduced with an ADA-expressing γ-retroviral vector without preconditioning or after 200 cGy or 900 cGy total-body irradiation and evaluated after 4 months. In all tissues analyzed, vector copy numbers (VCNs) were 100- to 1000-fold greater in mice receiving 900 cGy compared with 200 cGy (P < .05). In mice receiving 200 cGy, VCN was similar whether ERT was stopped or given for 1 or 4 months after GT. In unconditioned mice, there was decreased survival with and without ERT, and VCN was very low to undetectable. When recipients were conditioned with 200 cGy and received transduced lineage-depleted marrow, only recipients receiving ERT (1 or 4 months) had detectable vector sequences in thymocytes. In conclusion, cytoreduction is important for the engraftment of gene-transduced HSC, and short-term ERT after GT did not diminish the capacity of gene-corrected cells to engraft and persist.


Molecular Therapy | 2002

Expression from Second-Generation Feline Immunodeficiency Virus Vectors Is Impaired in Human Hematopoietic Cells

Mary A. Price; Scott S. Case; Denise A. Carbonaro; Xiao Jin Yu; Denise Petersen; Kathleen M. Sabo; Michael A. Curran; Barbara C. Engel; Hovanes Margarian; Janis L. Abkowitz; Garry P. Nolan; Donald B. Kohn

Vectors based on the feline immunodeficiency virus (FIV) have been developed as an alternative to those based on another lentivirus, human immunodeficiency virus-1 (HIV-1), because of theoretical safety advantages. We compared the efficiency of gene transfer and expression in human and feline hematopoietic progenitors using second-generation HIV-1 and FIV-based vectors. Vector pairs were tested using either human cytomegalovirus or murine phospho-glycerate kinase (PGK) internal promoters and were pseudotyped with the vesicular stomatitis virus G protein (VSV-G). Vector proviral copy numbers were similar in human and feline hematopoietic primary cells and cell lines transduced by HIV-1 or FIV vectors, demonstrating that both vectors are able to transfer genes efficiently to these cell types. HIV-1 vectors were well expressed in human primary hematopoietic cells and cell lines. However, transgene expression from FIV vectors was almost undetectable in human hematopoietic cells. In contrast, the FIV vector was expressed well in primary hematopoietic feline cells and human non-hematopoietic cells, demonstrating that low transgene expression from the FIV vector is a phenomenon specific to human hematopoietic cells. Northern blot analysis demonstrated decreased vector transcript levels in human CEM cells transduced with FIV relative to cells transduced with HIV-1, despite high vector copy numbers. No evidence of vector transcript instability was seen in studies of transduced CEM cells treated with actinomycin D. We conclude that FIV vectors can transfer genes into human hematopoietic cells as effectively as HIV-1 vectors, but that unknown elements in the current FIV backbone inhibit expression from FIV vectors in human hematopoietic cells.


Annals of the New York Academy of Sciences | 2006

Gene Therapy for Pediatric AIDS

Gerhard Bauer; David Selander; Barbara C. Engel; Denise A. Carbonaro; Susie Csik; Steve Rawlings; Joseph A. Church; Donald B. Kohn

Abstract: Gene therapy is an experimental treatment modality under investigation for applications to HIV‐1 infection. We have developed retroviral vectors carrying anti‐HIV‐1 genes, demonstrated that these genes cause significant suppression of HIV‐1 replication in cultures of primary hematopoietic cells, and performed a clinical trial in pediatric AIDS patients. Four HIV‐1‐infected children and adolescents underwent bone marrow harvest from which CD34+ cells were isolated and transduced by a retroviral vector carrying an RRE decoy gene. The cells were reinfused into the subjects, without complications, showing that gene transfer in pediatric AIDS patients is safe and feasible. However, gene‐containing leukocytes in the peripheral blood were seen only at a low level and only in the first months following cell infusion. To attain some degree of efficacy, it will be necessary to achieve a higher level of gene transfer and to obtain sustained gene expression. We are currently developing new gene transfer methods and vectors designed to improve the results in future trials. If it becomes possible to reach the ideal goal of producing high percentages of T lymphocytes and monocytic cells that are resistant to HIV‐1 infection, gene therapy could serve as a complement to antiretroviral drug therapy and help to sustain immunologic function.


Blood | 2008

Neonatal bone marrow transplantation of ADA-deficient SCID mice results in immunologic reconstitution despite low levels of engraftment and an absence of selective donor T lymphoid expansion.

Denise A. Carbonaro; Xiangyang Jin; Daniel Cotoi; Tiejuan Mi; Xiao Jin Yu; Dianne C. Skelton; Frederick J. Dorey; Rodney E. Kellems; Michael R. Blackburn; Donald B. Kohn

Adenosine deaminase (ADA)-deficient severe combined immune deficiency (SCID) may be treated by allogeneic hematopoietic stem cell transplantation without prior cytoreductive conditioning, although the mechanism of immune reconstitution is unclear. We studied this process in a murine gene knockout model of ADA-deficient SCID. Newborn ADA-deficient pups received transplants of intravenous infusion of normal congenic bone marrow, without prior cytoreductive conditioning, which resulted in long-term survival, multisystem correction, and nearly normal lymphocyte numbers and mitogenic proliferative responses. Only 1% to 3% of lymphocytes and myeloid cells were of donor origin without a selective expansion of donor-derived lymphocytes; immune reconstitution was by endogenous, host-derived ADA-deficient lymphocytes. Preconditioning of neonates with 100 to 400 cGy of total body irradiation before normal donor marrow transplant increased the levels of engrafted donor cells in a radiation dose-dependent manner, but the chimerism levels were similar for lymphoid and myeloid cells. The absence of selective reconstitution by donor T lymphocytes in the ADA-deficient mice indicates that restoration of immune function occurred by rescue of endogenous ADA-deficient lymphocytes through cross-correction from the engrafted ADA-replete donor cells. Thus, ADA-deficient SCID is unique in its responses to nonmyeloablative bone marrow transplantation, which has implications for clinical bone marrow transplantation or gene therapy.


Annals of the New York Academy of Sciences | 2003

Efficient characterization of retro-, lenti-, and foamyvector-transduced cell populations by high-accuracy insertion site sequencing

Manfred Schmidt; Hanno Glimm; Manuela Wissler; Gesa Hoffmann; Karin Olsson; Stephanie Sellers; Denise A. Carbonaro; John F. Tisdale; Cordula Leurs; Helmut Hanenberg; Cynthia E. Dunbar; Hans Peter Kiem; Stefan Karlsson; Donald B. Kohn; David M. Williams; Christof von Kalle

Abstract: The identification of unknown genomic flanking DNA sequences can be used for the molecular monitoring of retro‐, lenti‐ and foamyviral integration, transgenes in early embryogenesis, insertional mutagenesis, cell fate, and stem cell plasticity. Most existing methods reflect shortcomings in sensitivity and or specificity, thus limiting genomic sequencing of unknown flanking DNA to clonal preparations. The application of linear amplification‐mediated PCR (LAM‐PCR), a recently developed direct sequencing technique for flanking DNA, should circumvent current limitations in different research fields. This technique combines preamplification of target DNA with a unique succession of enzymatic reactions on solid‐phase. Using LAM‐PCR, we show the previously unfeasible in vivo retro‐, lenti‐ and foamyvirus integration site analysis in primate peripheral blood hematopoietic cells and human xenograft hematopoiesis. In light of two severe adverse events that occurred in a clinical SCID‐X1 gene therapy trial, in vivo monitoring of the reinfused transduced cell pool by integration site analysis will be an important component of each gene transfer and therapy study aimed at clinical use.


Journal of Clinical Investigation | 2017

Clinical efficacy of gene-modified stem cells in adenosine deaminase–deficient immunodeficiency

Kit L. Shaw; Elizabeth Garabedian; Suparna Mishra; Provaboti Barman; Alejandra Davila; Denise A. Carbonaro; Sally Shupien; Christopher Silvin; Sabine Geiger; Barbara Nowicki; E. Monika Smogorzewska; Berkley Brown; Xiaoyan Wang; Satiro N. De Oliveira; Yeong Choi; Alan K. Ikeda; Dayna Terrazas; Pei Yu Fu; Allen Yu; Beatriz Campo Fernandez; Aaron R. Cooper; Barbara C. Engel; Greg M. Podsakoff; Arumugam Balamurugan; Stacie M. Anderson; Linda M. Muul; G. Jayashree Jagadeesh; Neena Kapoor; John Tse; Theodore B. Moore

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) &ggr;-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.


Human Gene Therapy | 2017

Lentivirus mediated correction of artemis-deficient severe combined immunodeficiency

Divya Punwani; Misako Kawahara; Jason Yu; Ukina Sanford; Sushmita Roy; Kiran Patel; Denise A. Carbonaro; Andrea D. Karlen; Sara Khan; Kenneth Cornetta; Michael Rothe; Axel Schambach; Donald B. Kohn; Harry L. Malech; R. Scott McIvor; Jennifer M. Puck; Morton J. Cowan

During B and T lymphocyte maturation, V(D)J recombination is initiated by creation of DNA double-strand breaks. Artemis is an exonuclease essential for their subsequent repair by nonhomologous end-joining. Mutations in DCLRE1C, the gene encoding Artemis, cause T-B-NK+ severe combined immunodeficiency (ART-SCID) and also confer heightened sensitivity to ionizing radiation and alkylating chemotherapy. Although allogeneic hematopoietic cell transplantation can treat ART-SCID, conditioning regimens are poorly tolerated, leading to early mortality and/or late complications, including short stature, endocrinopathies, and dental aplasia. However, without alkylating chemotherapy as preconditioning, patients usually have graft rejection or limited T cell and no B cell recovery. Thus, addition of normal DCLRE1C cDNA to autologous hematopoietic stem cells is an attractive strategy to treat ART-SCID. We designed a self-inactivating lentivirus vector containing human Artemis cDNA under transcriptional regulation of the human endogenous Artemis promoter (AProArt). Fibroblasts from ART-SCID patients transduced with AProArt lentivirus showed correction of radiosensitivity. Mobilized peripheral blood CD34+ cells from an ART-SCID patient as well as hematopoietic stem cells from Artemis-deficient mice demonstrated restored T and B cell development following AProArt transduction. Murine hematopoietic cells transduced with AProArt exhibited no increase in replating potential in an in vitro immortalization assay, and analysis of AProArt lentivirus insertions showed no predilection for sites that could activate oncogenes. These efficacy and safety findings support institution of a clinical trial of gene addition therapy for ART-SCID.


Molecular Therapy | 2015

C-8. Immunological and Metabolic Correction After Lentiviral Vector Gene Therapy for ADA Deficiency

Hubert B. Gaspar; Karen F. Buckland; Denise A. Carbonaro; Kit L. Shaw; Provobati Barman; Alejandra Davila; Kimberly Gilmour; Claire Booth; Dayna Terrazs; Kenneth Cornetta; Anna Paruzynski; Manfred G. Schmidt; Robert A. Sokolic; Fabio Candotti; Adrian J. Thrasher; Donald B. Kohn

Background: Adenosine deaminase deficiency leads to severe combined immunodeficiency. Autologous haematopoietic stem cell gene therapy may offer a curative therapy. We developed a self inactivating lentiviral vector in which the human ADA gene is driven by an internal EFS promoter. Parallel trials using this vector were conducted in UCL, London and UCLA, Los AngelesObjective: A Phase I/II trial to assess the safety and efficacy of EFS-ADA lentiviral vector mediated gene modification of autologous CD34+ cells from ADA-deficient individuals.Methods: 20 patients (12 male; 8 female) aged between 0.4-6.5yrs were treated. All had been on enzyme replacement prior to treatment. CD34+ cells were collected either by BM harvest or peripheral blood stem cell mobilisation. Busulfan i.v. at a single dose of 4-5mg/kg was given as conditioning. CD34+ cells were stimulated with cytokines for 24hrs before transduction with vector for a further 18hrs. The dose of CD34+ cells returned ranged from 3-17 × 10e6/kg with a vector copy number (VCN) in the transduced population of between 0.25-6.3 vector copies per cell.Results: The procedure was well tolerated by all patients with no adverse events related to Busulfan conditioning or the vector. The follow up in all patients ranges from 1-35 months. All 20 patients remain off PEG ADA enzyme replacement therapy. There is evidence of immunological and metabolic recovery in all 16 patients treated for longer than 6 months. All patients have shown a rise in total T cell counts and at last follow-up the following levels were seen; total T cells, range 220-3,370; CD4+ cells 190-1509. Mitogen responses to PHA have increased in all patients and there is naive T cell recovery with an increase in CD45RA+CD4+CD27+ subpopulations and the number of TRECs. Immunoglobulin replacement therapy has been stopped in 4 patients so far and the one patient who has completed vaccinations shows normal vaccine specific responses. Gene marking is detectable in the periphery with highest marking in T cells but significant marking in B cells, NK cells and myeloid cells. Intracellular red blood cell ADA activity was negligible prior to gene therapy but at latest follow up is detectable at near or above normal levels in all patients. Integration site analysis shows occasional expansions but no persistence of expanded clones. There is no detection of expansion of clones with genes previously associated with insertional mutagenesis. All 20 patients are clinically well and the earliest treated patients are free of social restriction.Summary: Lentiviral vector mediated gene therapy for ADA deficiency is well tolerated and allows effective recovery of immunological and metabolic parameters with no evidence for mutagenesis thus far. These trials of 20 patients in two centres demonstrates the high efficacy and safety of this treatment strategy.

Collaboration


Dive into the Denise A. Carbonaro's collaboration.

Top Co-Authors

Avatar

Donald B. Kohn

University of California

View shared research outputs
Top Co-Authors

Avatar

Barbara C. Engel

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Fabio Candotti

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Gerhard Bauer

University of California

View shared research outputs
Top Co-Authors

Avatar

Kit L. Shaw

University of California

View shared research outputs
Top Co-Authors

Avatar

Xiangyang Jin

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ami J. Shah

University of California

View shared research outputs
Top Co-Authors

Avatar

Denise Petersen

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Elizabeth Garabedian

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge