151
|
Yamada K, Tsukahara T, Yoshino K, Kojima K, Agawa H, Yamashita Y, Amano Y, Hatta M, Matsuzaki Y, Kurotori N, Wakui K, Fukushima Y, Osada R, Shiozawa T, Sakashita K, Koike K, Kumaki S, Tanaka N, Takeshita T. Identification of a high incidence region for retroviral vector integration near exon 1 of the LMO2 locus. Retrovirology 2009; 6:79. [PMID: 19725963 PMCID: PMC2742512 DOI: 10.1186/1742-4690-6-79] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Accepted: 09/02/2009] [Indexed: 01/19/2023] Open
Abstract
Therapeutic retroviral vector integration near the oncogene LMO2 is thought to be a cause of leukemia in X-SCID gene therapy trials. However, no published studies have evaluated the frequency of vector integrations near exon 1 of the LMO2 locus. We identified a high incidence region (HIR) of vector integration using PCR techniques in the upstream region close to the LMO2 transcription start site in the TPA-Mat T cell line. The integration frequency of the HIR was one per 4.46 x 10(4) cells. This HIR was also found in Jurkat T cells but was absent from HeLa cells. Furthermore, using human cord blood-derived CD34+ cells we identified a HIR in a similar region as the TPA-Mat T cell line. One of the X-linked severe combined immunodeficiency (X-SCID) patients that developed leukemia after gene therapy had a vector integration site in this HIR. Therefore, the descriptions of the location and the integration frequency of the HIR presented here may help us to better understand vector-induced leukemogenesis.
Collapse
Affiliation(s)
- Koichiro Yamada
- Department of Microbiology and Immunology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
152
|
Gaide O. Gene therapy and protein therapy of ectodermal dysplasias: A perspective. Am J Med Genet A 2009; 149A:2042-4. [DOI: 10.1002/ajmg.a.32839] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
153
|
Ng CEL, Ito Y, Osato M. Retroviral integration sites (RIS) mark cis-regulatory elements. Crit Rev Oncol Hematol 2009; 71:1-11. [DOI: 10.1016/j.critrevonc.2008.10.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2008] [Revised: 09/29/2008] [Accepted: 10/15/2008] [Indexed: 12/15/2022] Open
|
154
|
Kustikova OS, Schiedlmeier B, Brugman MH, Stahlhut M, Bartels S, Li Z, Baum C. Cell-intrinsic and vector-related properties cooperate to determine the incidence and consequences of insertional mutagenesis. Mol Ther 2009; 17:1537-47. [PMID: 19532134 DOI: 10.1038/mt.2009.134] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
In gene therapeutic approaches targeting hematopoietic cells, insertional mutagenesis may provoke clonal dominance with potential progress to overt leukemia. To investigate the contribution of cell-intrinsic features and determine the frequency of insertional proto-oncogene activation, we sorted hematopoietic subpopulations before transduction with replication-deficient gamma-retroviral vectors and studied the clonal repertoire in transplanted C57BL/6J mice. Progressive clonal dominance only developed in the progeny of populations with intrinsic stem cell potential, where expanding clones with insertional upregulation of proto-oncogenes such as Evi1 were retrieved with a frequency of approximately 10(-4). Longitudinal studies by high-throughput sequencing and locus-specific quantitative PCR showed clones with >50-fold expansion between weeks 5 and 31 after transplantation. In contrast, insertional events in proto-oncogenes did not endow the progeny of multipotent or myeloid-restricted progenitors with the potential for clonal dominance (risk <10(-6)). Transducing sorted hematopoietic stem cells (HSCs) with self-inactivating (SIN) lentiviral vectors in short-term cultures improved chimerism, and although clonal dominance developed, there was no evidence for insertional events in the vicinity of proto-oncogenes as the underlying cause. We conclude that cell-intrinsic properties cooperate with vector-related features to determine the incidence and consequences of insertional mutagenesis. Furthermore, our study offers perspectives for refinement of animal experiments in the assessment of vector-related genotoxicity.
Collapse
Affiliation(s)
- Olga S Kustikova
- Department of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
155
|
Liuba K, Pronk CJH, Stott SRW, Jacobsen SEW. Polyclonal T-cell reconstitution of X-SCID recipients after in utero transplantation of lymphoid-primed multipotent progenitors. Blood 2009; 113:4790-8. [PMID: 19074736 DOI: 10.1182/blood-2007-12-129056] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Although successful in utero hematopoietic cell transplantation (IUHCT) of X-linked severe combined immune deficiency (X-SCID) with enriched stem and progenitor cells was achieved more than a decade ago, it remains applied only in rare cases. Although this in part reflects that postnatal transplantations have overall given good results, there are no direct comparisons between IUHCT and postnatal transplantations of X-SCID. The proposed tolerance of the fetal immune system to foreign human leukocyte antigen early in gestation, a main rationale behind IUHCT, has recently been challenged by evidence for a considerable immune barrier against in utero transplanted allogeneic bone marrow cells. Consequently, there is need for further exploring the application of purified stem and progenitor cells to overcome this barrier also in IUHCT. Herein, we demonstrate in a congenic setting that recently identified lymphoid-primed multipotent progenitors are superior to hematopoietic stem cells in providing rapid lymphoid reconstitution after IUHCT of X-SCID recipients, and sustain in the long-term B cells, polyclonal T cells, as well as short-lived B-cell progenitors and thymic T-cell precursors. We further provide evidence for IUHCT of hematopoietic stem cells giving superior B- and T-cell reconstitution in fetal X-SCID recipients compared with neonatal and adolescent recipients.
Collapse
Affiliation(s)
- Karina Liuba
- Hematopoietic Stem Cell Laboratory, Lund Strategic Research Center for Stem Cell, Biology and Cell Therapy, Lund University, Lund, Sweden
| | | | | | | |
Collapse
|
156
|
Wang GP, Levine BL, Binder GK, Berry CC, Malani N, McGarrity G, Tebas P, June CH, Bushman FD. Analysis of lentiviral vector integration in HIV+ study subjects receiving autologous infusions of gene modified CD4+ T cells. Mol Ther 2009; 17:844-50. [PMID: 19259065 PMCID: PMC2835137 DOI: 10.1038/mt.2009.16] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Accepted: 01/12/2009] [Indexed: 12/18/2022] Open
Abstract
Lentiviral vector-based gene therapy has been used to target the human immunodeficiency virus (HIV) using an antisense env payload. We have analyzed lentiviral-vector integration sites from three treated individuals. We compared integration sites from the ex vivo vector-transduced CD4+ cell products to sites from cells recovered at several times after infusion. Integration sites were analyzed using 454 pyrosequencing, yielding a total of 7,782 unique integration sites from the ex vivo product and 237 unique sites from cells recovered after infusion. Integrated vector copies in both data sets were found to be strongly enriched within active genes and near epigenetic marks associated with active transcription units. Analysis of integration relative to nucleosome structure on target DNA indicated favoring of integration in outward facing DNA major grooves on the nucleosome surface. There was no indication that growth of transduced cells after infusion resulted in enrichment for integration sites near proto-oncogene 5'-ends or within tumor suppressor genes. Thus, this first look at the longitudinal evolution of cells transduced with a lentiviral vector after infusion of gene modified CD4+ cells provided no evidence for abnormal expansions of cells due to vector-mediated insertional activation of proto-oncogenes.
Collapse
Affiliation(s)
- Gary P Wang
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-6076, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
157
|
Davé UP, Akagi K, Tripathi R, Cleveland SM, Thompson MA, Yi M, Stephens R, Downing JR, Jenkins NA, Copeland NG. Murine leukemias with retroviral insertions at Lmo2 are predictive of the leukemias induced in SCID-X1 patients following retroviral gene therapy. PLoS Genet 2009; 5:e1000491. [PMID: 19461887 PMCID: PMC2679194 DOI: 10.1371/journal.pgen.1000491] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Accepted: 04/22/2009] [Indexed: 01/03/2023] Open
Abstract
Five X-linked severe combined immunodeficiency patients (SCID-X1) successfully treated with autologous bone marrow stem cells infected ex vivo with an IL2RG-containing retrovirus subsequently developed T-cell leukemia and four contained insertional mutations at LMO2. Genetic evidence also suggests a role for IL2RG in tumor formation, although this remains controversial. Here, we show that the genes and signaling pathways deregulated in murine leukemias with retroviral insertions at Lmo2 are similar to those deregulated in human leukemias with high LMO2 expression and are highly predictive of the leukemias induced in SCID-X1 patients. We also provide additional evidence supporting the notion that IL2RG and LMO2 cooperate in leukemia induction but are not sufficient and require additional cooperating mutations. The highly concordant nature of the genetic events giving rise to mouse and human leukemias with mutations at Lmo2 are an encouraging sign to those wanting to use mice to model human cancer and may help in designing safer methods for retroviral gene therapy.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Animals
- Base Sequence
- DNA, Neoplasm/genetics
- DNA-Binding Proteins/genetics
- Genetic Therapy/adverse effects
- Hematopoietic Stem Cell Transplantation/adverse effects
- Humans
- Interleukin Receptor Common gamma Subunit/genetics
- LIM Domain Proteins
- Leukemia, Experimental/etiology
- Leukemia, Experimental/genetics
- Leukemia, Experimental/pathology
- Leukemia-Lymphoma, Adult T-Cell/etiology
- Leukemia-Lymphoma, Adult T-Cell/genetics
- Leukemia-Lymphoma, Adult T-Cell/pathology
- Metalloproteins/genetics
- Mice
- Mice, SCID
- Models, Genetic
- Molecular Sequence Data
- Mutagenesis, Insertional
- Proto-Oncogene Proteins
- Retroviridae/genetics
- Transplantation, Autologous
- Virus Integration/genetics
- X-Linked Combined Immunodeficiency Diseases/complications
- X-Linked Combined Immunodeficiency Diseases/genetics
- X-Linked Combined Immunodeficiency Diseases/therapy
Collapse
Affiliation(s)
- Utpal P Davé
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
158
|
Maruggi G, Porcellini S, Facchini G, Perna SK, Cattoglio C, Sartori D, Ambrosi A, Schambach A, Baum C, Bonini C, Bovolenta C, Mavilio F, Recchia A. Transcriptional enhancers induce insertional gene deregulation independently from the vector type and design. Mol Ther 2009; 17:851-6. [PMID: 19293778 PMCID: PMC2835143 DOI: 10.1038/mt.2009.51] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Accepted: 02/22/2009] [Indexed: 11/10/2022] Open
Abstract
The integration characteristics of retroviral (RV) vectors increase the probability of interfering with the regulation of cellular genes, and account for a tangible risk of insertional mutagenesis in treated patients. To assess the potential genotoxic risk of conventional or self-inactivating (SIN) gamma-RV and lentiviral (LV) vectors independently from the biological consequences of the insertion event, we developed a quantitative assay based on real-time reverse transcriptase--PCR on low-density arrays to evaluate alterations of gene expression in individual primary T-cell clones. We show that the Moloney leukemia virus long terminal repeat (LTR) enhancer has the strongest activity in both a gamma-RV and a LV vector context, while an internal cellular promoter induces deregulation of gene expression less frequently, at a shorter range and to a lower extent in both vector types. Downregulation of gene expression was observed only in the context of LV vectors. This study indicates that insertional gene activation is determined by the characteristics of the transcriptional regulatory elements carried by the vector, and is largely independent from the vector type or design.
Collapse
Affiliation(s)
- Giulietta Maruggi
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
159
|
Modlich U, Baum C. Preventing and exploiting the oncogenic potential of integrating gene vectors. J Clin Invest 2009; 119:755-8. [PMID: 19348042 DOI: 10.1172/jci38831] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Gene therapy requires efficient gene delivery to cure or prevent disease by modifying the genome of somatic cells. However, gene vectors, which insert themselves into the host genome in order to achieve persistent protein expression, can trigger oncogenesis by upregulating cellular protooncogenes. This adverse event, known as insertional mutagenesis, has become a major hurdle in the field. Vectors developed on the basis of lentiviruses are considered to be less genotoxic than the hitherto used gamma-retroviral vectors. For their report in this issue of the JCI, Montini et al. utilized a tumor-prone mouse model to identify the genetic determinants of insertional mutagenesis (see the related article beginning on page 964). They report that the lentiviral integration pattern and additional improvements in vector design reduce the genotoxic risk. These findings will inform future vector design with the goal of limiting genotoxicity for gene therapy or increasing genotoxicity for protooncogene discovery.
Collapse
Affiliation(s)
- Ute Modlich
- Department of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | | |
Collapse
|
160
|
QuickMap: a public tool for large-scale gene therapy vector insertion site mapping and analysis. Gene Ther 2009; 16:885-93. [PMID: 19387483 DOI: 10.1038/gt.2009.37] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Several events of insertional mutagenesis in pre-clinical and clinical gene therapy studies have created intense interest in assessing the genomic insertion profiles of gene therapy vectors. For the construction of such profiles, vector-flanking sequences detected by inverse PCR, linear amplification-mediated-PCR or ligation-mediated-PCR need to be mapped to the host cell's genome and compared to a reference set. Although remarkable progress has been achieved in mapping gene therapy vector insertion sites, public reference sets are lacking, as are the possibilities to quickly detect non-random patterns in experimental data. We developed a tool termed QuickMap, which uniformly maps and analyzes human and murine vector-flanking sequences within seconds (available at www.gtsg.org). Besides information about hits in chromosomes and fragile sites, QuickMap automatically determines insertion frequencies in +/- 250 kb adjacency to genes, cancer genes, pseudogenes, transcription factor and (post-transcriptional) miRNA binding sites, CpG islands and repetitive elements (short interspersed nuclear elements (SINE), long interspersed nuclear elements (LINE), Type II elements and LTR elements). Additionally, all experimental frequencies are compared with the data obtained from a reference set, containing 1 000 000 random integrations ('random set'). Thus, for the first time a tool allowing high-throughput profiling of gene therapy vector insertion sites is available. It provides a basis for large-scale insertion site analyses, which is now urgently needed to discover novel gene therapy vectors with 'safe' insertion profiles.
Collapse
|
161
|
Montini E, Cesana D, Schmidt M, Sanvito F, Bartholomae CC, Ranzani M, Benedicenti F, Sergi LS, Ambrosi A, Ponzoni M, Doglioni C, Di Serio C, von Kalle C, Naldini L. The genotoxic potential of retroviral vectors is strongly modulated by vector design and integration site selection in a mouse model of HSC gene therapy. J Clin Invest 2009; 119:964-75. [PMID: 19307726 PMCID: PMC2662564 DOI: 10.1172/jci37630] [Citation(s) in RCA: 426] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Accepted: 01/14/2009] [Indexed: 12/25/2022] Open
Abstract
gamma-Retroviral vectors (gammaRVs), which are commonly used in gene therapy, can trigger oncogenesis by insertional mutagenesis. Here, we have dissected the contribution of vector design and viral integration site selection (ISS) to oncogenesis using an in vivo genotoxicity assay based on transplantation of vector-transduced tumor-prone mouse hematopoietic stem/progenitor cells. By swapping genetic elements between gammaRV and lentiviral vectors (LVs), we have demonstrated that transcriptionally active long terminal repeats (LTRs) are major determinants of genotoxicity even when reconstituted in LVs and that self-inactivating (SIN) LTRs enhance the safety of gammaRVs. By comparing the genotoxicity of vectors with matched active LTRs, we were able to determine that substantially greater LV integration loads are required to approach the same oncogenic risk as gammaRVs. This difference in facilitating oncogenesis is likely to be explained by the observed preferential targeting of cancer genes by gammaRVs. This integration-site bias was intrinsic to gammaRVs, as it was also observed for SIN gammaRVs that lacked genotoxicity in our model. Our findings strongly support the use of SIN viral vector platforms and show that ISS can substantially modulate genotoxicity.
Collapse
Affiliation(s)
- Eugenio Montini
- San Raffaele-Telethon Institute for Gene Therapy and
Vita Salute San Raffaele University, Milan, Italy.
National Center for Tumor Diseases, Heidelberg, Germany.
Department of Pathology, San Raffaele Hospital, Milan, Italy.
University Centre of Statistics for Biomedical Sciences, Vita Salute San Raffaele University, Milan, Italy
| | - Daniela Cesana
- San Raffaele-Telethon Institute for Gene Therapy and
Vita Salute San Raffaele University, Milan, Italy.
National Center for Tumor Diseases, Heidelberg, Germany.
Department of Pathology, San Raffaele Hospital, Milan, Italy.
University Centre of Statistics for Biomedical Sciences, Vita Salute San Raffaele University, Milan, Italy
| | - Manfred Schmidt
- San Raffaele-Telethon Institute for Gene Therapy and
Vita Salute San Raffaele University, Milan, Italy.
National Center for Tumor Diseases, Heidelberg, Germany.
Department of Pathology, San Raffaele Hospital, Milan, Italy.
University Centre of Statistics for Biomedical Sciences, Vita Salute San Raffaele University, Milan, Italy
| | - Francesca Sanvito
- San Raffaele-Telethon Institute for Gene Therapy and
Vita Salute San Raffaele University, Milan, Italy.
National Center for Tumor Diseases, Heidelberg, Germany.
Department of Pathology, San Raffaele Hospital, Milan, Italy.
University Centre of Statistics for Biomedical Sciences, Vita Salute San Raffaele University, Milan, Italy
| | - Cynthia C. Bartholomae
- San Raffaele-Telethon Institute for Gene Therapy and
Vita Salute San Raffaele University, Milan, Italy.
National Center for Tumor Diseases, Heidelberg, Germany.
Department of Pathology, San Raffaele Hospital, Milan, Italy.
University Centre of Statistics for Biomedical Sciences, Vita Salute San Raffaele University, Milan, Italy
| | - Marco Ranzani
- San Raffaele-Telethon Institute for Gene Therapy and
Vita Salute San Raffaele University, Milan, Italy.
National Center for Tumor Diseases, Heidelberg, Germany.
Department of Pathology, San Raffaele Hospital, Milan, Italy.
University Centre of Statistics for Biomedical Sciences, Vita Salute San Raffaele University, Milan, Italy
| | - Fabrizio Benedicenti
- San Raffaele-Telethon Institute for Gene Therapy and
Vita Salute San Raffaele University, Milan, Italy.
National Center for Tumor Diseases, Heidelberg, Germany.
Department of Pathology, San Raffaele Hospital, Milan, Italy.
University Centre of Statistics for Biomedical Sciences, Vita Salute San Raffaele University, Milan, Italy
| | - Lucia Sergi Sergi
- San Raffaele-Telethon Institute for Gene Therapy and
Vita Salute San Raffaele University, Milan, Italy.
National Center for Tumor Diseases, Heidelberg, Germany.
Department of Pathology, San Raffaele Hospital, Milan, Italy.
University Centre of Statistics for Biomedical Sciences, Vita Salute San Raffaele University, Milan, Italy
| | - Alessandro Ambrosi
- San Raffaele-Telethon Institute for Gene Therapy and
Vita Salute San Raffaele University, Milan, Italy.
National Center for Tumor Diseases, Heidelberg, Germany.
Department of Pathology, San Raffaele Hospital, Milan, Italy.
University Centre of Statistics for Biomedical Sciences, Vita Salute San Raffaele University, Milan, Italy
| | - Maurilio Ponzoni
- San Raffaele-Telethon Institute for Gene Therapy and
Vita Salute San Raffaele University, Milan, Italy.
National Center for Tumor Diseases, Heidelberg, Germany.
Department of Pathology, San Raffaele Hospital, Milan, Italy.
University Centre of Statistics for Biomedical Sciences, Vita Salute San Raffaele University, Milan, Italy
| | - Claudio Doglioni
- San Raffaele-Telethon Institute for Gene Therapy and
Vita Salute San Raffaele University, Milan, Italy.
National Center for Tumor Diseases, Heidelberg, Germany.
Department of Pathology, San Raffaele Hospital, Milan, Italy.
University Centre of Statistics for Biomedical Sciences, Vita Salute San Raffaele University, Milan, Italy
| | - Clelia Di Serio
- San Raffaele-Telethon Institute for Gene Therapy and
Vita Salute San Raffaele University, Milan, Italy.
National Center for Tumor Diseases, Heidelberg, Germany.
Department of Pathology, San Raffaele Hospital, Milan, Italy.
University Centre of Statistics for Biomedical Sciences, Vita Salute San Raffaele University, Milan, Italy
| | - Christof von Kalle
- San Raffaele-Telethon Institute for Gene Therapy and
Vita Salute San Raffaele University, Milan, Italy.
National Center for Tumor Diseases, Heidelberg, Germany.
Department of Pathology, San Raffaele Hospital, Milan, Italy.
University Centre of Statistics for Biomedical Sciences, Vita Salute San Raffaele University, Milan, Italy
| | - Luigi Naldini
- San Raffaele-Telethon Institute for Gene Therapy and
Vita Salute San Raffaele University, Milan, Italy.
National Center for Tumor Diseases, Heidelberg, Germany.
Department of Pathology, San Raffaele Hospital, Milan, Italy.
University Centre of Statistics for Biomedical Sciences, Vita Salute San Raffaele University, Milan, Italy
| |
Collapse
|
162
|
Sustained high-level polyclonal hematopoietic marking and transgene expression 4 years after autologous transplantation of rhesus macaques with SIV lentiviral vector-transduced CD34+ cells. Blood 2009; 113:5434-43. [PMID: 19339698 DOI: 10.1182/blood-2008-10-185199] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We previously reported that lentiviral vectors derived from the simian immunodeficiency virus (SIV) were efficient at transducing rhesus hematopoietic repopulating cells. To evaluate the persistence of vector-containing and -expressing cells long term, and the safety implications of SIV lentiviral vector-mediated gene transfer, we followed 3 rhesus macaques for more than 4 years after transplantation with transduced CD34+ cells. All 3 animals demonstrated significant vector marking and expression of the GFP transgene in T cells, B cells, and granulocytes, with mean GFP+ levels of 6.7% (range, 3.3%-13.0%), 7.4% (4.2%-13.4%), and 5.6% (3.1%-10.5%), respectively. There was no vector silencing in hematopoietic cells over time. Vector insertion site analysis of granulocytes demonstrated sustained highly polyclonal reconstitution, with no evidence for progression to oligoclonality. A significant number of clones were found to contribute at both 1-year and 3- or 4-year time points. No vector integrations were detected in the MDS1/EVI1 region, in contrast to our previous findings with a gamma-retroviral vector. These data show that lentiviral vectors can mediate stable and efficient long-term expression in the progeny of transduced hematopoietic stem cells, with an integration profile that may be safer than that of standard Moloney murine leukemia virus (MLV)-derived retroviral vectors.
Collapse
|
163
|
Scobie L, Hector RD, Grant L, Bell M, Nielsen AA, Meikle S, Philbey A, Philbey A, Thrasher AJ, Thrasher AJ, Cameron ER, Blyth K, Neil JC. A novel model of SCID-X1 reconstitution reveals predisposition to retrovirus-induced lymphoma but no evidence of gammaC gene oncogenicity. Mol Ther 2009; 17:1031-8. [PMID: 19337236 DOI: 10.1038/mt.2009.59] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The emergence of leukemia following gene transfer to restore common cytokine receptor gamma chain (gammaC) function in X-linked severe combined immunodeficiency (SCID-X1) has raised important questions with respect to gene therapy safety. To explore the risk factors involved, we tested the oncogenic potential of human gammaC in new strains of transgenic mice expressing the gene under the control of the CD2 promoter and locus control region (LCR). These mice demonstrated mildly perturbed T-cell development, with an increased proportion of thymic CD8 cells, but showed no predisposition to tumor development even on highly tumor prone backgrounds or after gamma-retrovirus infection. The human CD2-gammaC transgene rescued T and B-cell development in gammaC(-/-) mice but with an age-related delay, mimicking postnatal reconstitution in SCID-X1 gene therapy subjects. However, we noted that gammaC(-/-) mice are acutely susceptible to murine leukemia virus (MLV) leukemogenesis, and that this trait was not corrected by the gammaC transgene. We conclude that the SCID-X1 phenotype can be corrected safely by stable ectopic expression of gammaC and that the transgene is not significantly oncogenic when expressed in this context. However, an underlying predisposition conferred by the SCID-X1 background appears to collaborate with insertional mutagenesis to increase the risk of tumor development.
Collapse
Affiliation(s)
- Linda Scobie
- Division of Pathological Sciences, Institute of Comparative Medicine, Faculty of Veterinary Medicine, University of Glasgow, Glasgow, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
164
|
Cancer gene discovery in mouse and man. Biochim Biophys Acta Rev Cancer 2009; 1796:140-61. [PMID: 19285540 PMCID: PMC2756404 DOI: 10.1016/j.bbcan.2009.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2009] [Revised: 03/03/2009] [Accepted: 03/05/2009] [Indexed: 12/31/2022]
Abstract
The elucidation of the human and mouse genome sequence and developments in high-throughput genome analysis, and in computational tools, have made it possible to profile entire cancer genomes. In parallel with these advances mouse models of cancer have evolved into a powerful tool for cancer gene discovery. Here we discuss the approaches that may be used for cancer gene identification in both human and mouse and discuss how a cross-species 'oncogenomics' approach to cancer gene discovery represents a powerful strategy for finding genes that drive tumourigenesis.
Collapse
|
165
|
Felice B, Cattoglio C, Cittaro D, Testa A, Miccio A, Ferrari G, Luzi L, Recchia A, Mavilio F. Transcription factor binding sites are genetic determinants of retroviral integration in the human genome. PLoS One 2009; 4:e4571. [PMID: 19238208 PMCID: PMC2642719 DOI: 10.1371/journal.pone.0004571] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2008] [Accepted: 01/16/2009] [Indexed: 01/10/2023] Open
Abstract
Gamma-retroviruses and lentiviruses integrate non-randomly in mammalian genomes, with specific preferences for active chromatin, promoters and regulatory regions. Gene transfer vectors derived from gamma-retroviruses target at high frequency genes involved in the control of growth, development and differentiation of the target cell, and may induce insertional tumors or pre-neoplastic clonal expansions in patients treated by gene therapy. The gene expression program of the target cell is apparently instrumental in directing gamma-retroviral integration, although the molecular basis of this phenomenon is poorly understood. We report a bioinformatic analysis of the distribution of transcription factor binding sites (TFBSs) flanking >4,000 integrated proviruses in human hematopoietic and non-hematopoietic cells. We show that gamma-retroviral, but not lentiviral vectors, integrate in genomic regions enriched in cell-type specific subsets of TFBSs, independently from their relative position with respect to genes and transcription start sites. Analysis of sequences flanking the integration sites of Moloney leukemia virus (MLV)- and human immunodeficiency virus (HIV)-derived vectors carrying mutations in their long terminal repeats (LTRs), and of HIV vectors packaged with an MLV integrase, indicates that the MLV integrase and LTR enhancer are the viral determinants of the selection of TFBS-rich regions in the genome. This study identifies TFBSs as differential genomic determinants of retroviral target site selection in the human genome, and suggests that transcription factors binding the LTR enhancer may synergize with the integrase in tethering retroviral pre-integration complexes to transcriptionally active regulatory regions. Our data indicate that gamma-retroviruses and lentiviruses have evolved dramatically different strategies to interact with the host cell chromatin, and predict a higher risk in using gamma-retroviral vs. lentiviral vectors for human gene therapy applications.
Collapse
Affiliation(s)
- Barbara Felice
- IFOM, FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Claudia Cattoglio
- IIT Unit of Molecular Neuroscience, Istituto Scientifico H. San Raffaele, Milan, Italy
| | - Davide Cittaro
- Cogentech, Consortium for Genomic Technologies, Milan, Italy
| | - Anna Testa
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Giuliana Ferrari
- HSR-Telethon Institute of Gene Therapy, Milan, Italy
- Vita-Salute University, Milan, Italy
| | - Lucilla Luzi
- IFOM, FIRC Institute of Molecular Oncology Foundation, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Alessandra Recchia
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Fulvio Mavilio
- IIT Unit of Molecular Neuroscience, Istituto Scientifico H. San Raffaele, Milan, Italy
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
166
|
Hayakawa J, Washington K, Uchida N, Phang O, Kang EM, Hsieh MM, Tisdale JF. Long-term vector integration site analysis following retroviral mediated gene transfer to hematopoietic stem cells for the treatment of HIV infection. PLoS One 2009; 4:e4211. [PMID: 19148292 PMCID: PMC2615408 DOI: 10.1371/journal.pone.0004211] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Accepted: 11/25/2008] [Indexed: 12/24/2022] Open
Abstract
We previously reported the efficacy of nonmyeloablative allogeneic transplantation in 2 HIV positive recipients, one of whom received retrovirus transduced hematopoietic stem cells to confer resistance to HIV. Here we report an assessment of retroviral integration sites (RISs) recovered out to 3 years post-transplantation. We identified 213 unique RISs from the patient's peripheral blood samples by linear amplification-mediated PCR (LAM-PCR). While vector integration patterns were similar to that previously reported, only 3.76% of RISs were common among early (up to 3 months) and late samples (beyond 1 year). Additionally, common integration sites were enriched among late samples (14.9% vs. 36.8%, respectively). Three RISs were found near or within known oncogenes, but 2 were limited to early timepoints. Interestingly, an integration site near the MDS1 gene was detected in long-term follow-up samples; however, the overall contribution of MDS1 integrated clone remained stably low during follow-up.
Collapse
Affiliation(s)
- Jun Hayakawa
- Molecular and Clinical Hematology Branch (MCHB), National Institutes of Diabetes and Digestive and Kidney Disorders (NIDDK) and National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Kareem Washington
- Molecular and Clinical Hematology Branch (MCHB), National Institutes of Diabetes and Digestive and Kidney Disorders (NIDDK) and National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Naoya Uchida
- Molecular and Clinical Hematology Branch (MCHB), National Institutes of Diabetes and Digestive and Kidney Disorders (NIDDK) and National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Oswald Phang
- Molecular and Clinical Hematology Branch (MCHB), National Institutes of Diabetes and Digestive and Kidney Disorders (NIDDK) and National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Elizabeth M. Kang
- Molecular and Clinical Hematology Branch (MCHB), National Institutes of Diabetes and Digestive and Kidney Disorders (NIDDK) and National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Matthew M. Hsieh
- Molecular and Clinical Hematology Branch (MCHB), National Institutes of Diabetes and Digestive and Kidney Disorders (NIDDK) and National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - John F. Tisdale
- Molecular and Clinical Hematology Branch (MCHB), National Institutes of Diabetes and Digestive and Kidney Disorders (NIDDK) and National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
167
|
Abstract
Dose-limiting toxicity of chemotherapeutic agents, i.e., myelosuppression, can limit their effectiveness. The transfer and expression of drug-resistance genes might decrease the risks associated with acute hematopoietic toxicity. Protection of hematopoietic stem/progenitor cells by transfer of drug-resistance genes provides the possibility of intensification or escalation of antitumor drug doses and consequently an improved therapeutic index. This chapter reviews drug-resistance gene transfer strategies for either myeloprotection or therapeutic gene selection. Selecting candidate drug-resistance gene(s), gene transfer methodology, evaluating the safety and the efficiency of the treatment strategy, relevant in vivo models, and oncoretroviral transduction of human hematopoietic stem/progenitor cells under clinically applicable conditions are described.
Collapse
Affiliation(s)
- Tulin Budak-Alpdogan
- Department of Medicine, The Cancer Institute of New Jersey, Robert Wood Johson Medical School, University of Medicine & Dentistry of New Jersey, New Brunswick, NJ, USA
| | | |
Collapse
|
168
|
Schmidt M, Schwarzwaelder K, Bartholomae CC, Glimm H, von Kalle C. Detection of retroviral integration sites by linear amplification-mediated PCR and tracking of individual integration clones in different samples. Methods Mol Biol 2009; 506:363-72. [PMID: 19110638 DOI: 10.1007/978-1-59745-409-4_24] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In order to restore or to introduce a gene function integrating viral vector systems are used to genetically modify hematopoietic stem cells. The occurrence of immortalized cell clones after transduction in vitro (Blood 106:3932-3939, 2005) and clonal dominance as well as leukemia in preclinical (Nat. Med. 12:401- 409, 2006; Blood 106:2530-2533, 2005; Science 308:1171-1174, 2005; Science 296:497, 2002; Blood 107:3865-3867, 2006) and clinical (Nat. Med. 12:401-409, 2006; Science 302:415-419, 2003; J. Clin. Invest. 118:3143-3150, 2008) gene therapy trials revealed that the nondirected integration of a vector may be associated with serious side effects. By means of the linear amplification-mediated PCR (LAM-PCR) (Blood 100:2737-2743, 2002; Nat. Methods 4:1051-1057, 2007) it is possible to identify miscellaneous vector-genome junctions in one sample, each unique for one integration clone down to the single cell level. Thus this method allows to determine the clonality of a genetically modified hematopoietic repopulation as well as to sequence the vector integration sites and therefore to analyze the integration site distribution and the influence of the vector integration site on the cell fate. The recognition of the integration site sequence corresponding to a specific clone allows the tracking of an individual clone in various samples.
Collapse
Affiliation(s)
- Manfred Schmidt
- Department of Translational Oncology, National Center of Tumor Diseases, Heidelberg, Germany
| | | | | | | | | |
Collapse
|
169
|
Cornils K, Lange C, Schambach A, Brugman MH, Nowak R, Lioznov M, Baum C, Fehse B. Stem cell marking with promotor-deprived self-inactivating retroviral vectors does not lead to induced clonal imbalance. Mol Ther 2009; 17:131-43. [PMID: 19002163 PMCID: PMC2834973 DOI: 10.1038/mt.2008.238] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2008] [Accepted: 09/30/2008] [Indexed: 12/19/2022] Open
Abstract
Stable genetic modification of stem cells holds great promise for gene therapy and marking, but commonly used gamma-retroviral vectors were found to influence growth/survival characteristics of hematopoietic stem cells (HSCs) by insertional mutagenesis. In this article, we show that promoter-deprived gamma-retroviral self-inactivating (pd-SIN) vectors allow stable genetic marking of serially reconstituting murine HSC. In contrast to findings with gamma-retroviral long terminal repeat (LTR) vectors, serial transplantation of pd-SIN-marked HSC in a sensitive mouse model was apparently not associated with induced clonal imbalance of gene-marked HSC. Furthermore, insertions of pd-SIN into protooncogenes, growth-promoting and signaling genes occurred significantly less frequent than in control experiments with LTR vectors. Also, transcriptional dysregulation of neighboring genes potentially caused by the pd-SIN insertion was rarely seen and comparatively weak. The integration pattern of promotor-deprived SIN vectors in reconstituting HSC seems to depend on the transcriptional activity of the respective gene loci reflecting the picture described for LTR vectors. In conclusion, our data strongly support the use of SIN vectors for gene-marking studies and suggest an increased therapeutic index for vectors lacking enhancers active in HSC.
Collapse
Affiliation(s)
- Kerstin Cornils
- Experimental Pediatric Oncology and Hematology, Pediatric Clinic III, University Hospital of the Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | | | | | | | | | | | | | | |
Collapse
|
170
|
DiMichele DM. Ethical considerations in clinical investigation: exploring relevance in haemophilia research. Haemophilia 2008; 14 Suppl 3:122-9. [PMID: 18510532 DOI: 10.1111/j.1365-2516.2008.01738.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Painful controversy has so far been largely absent from the history of haemophilia-related clinical research. However, the investigative methods now needed to realize evidence-based clinical practice, therapeutic advance, and a progressive standard of care for patients worldwide will be accompanied by the potential for ethical dilemma and transgression. From the current vantage point, three primary ethical issues merit special consideration: (i) the therapeutic misconception inherent to all clinical research and the randomized trial in particular; (ii) high risk and potentially non-beneficial novel technology research in children; and (iii) a collaborative partnership approach to research in the developing world. This study will focus on a discussion of each of these, drawing from the research ethics literature to offer a potential template for future deliberations in clinical trial design.
Collapse
Affiliation(s)
- D M DiMichele
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
171
|
Genome areas with high gene density and CpG island neighborhood strongly attract porcine endogenous retrovirus for integration and favor the formation of hot spots. J Virol 2008; 83:1920-9. [PMID: 19036816 DOI: 10.1128/jvi.00856-08] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Porcine endogenous retroviruses (PERV) are members of the gammaretrovirus genus and display integration preferences around transcription start sites, a finding which is similar to the preferences of the murine leukemia virus (MLV). Our new genome-wide analysis of the integration profile of a recombinant PERV (PERV A/C), enabled us to examine more than 1,900 integration sites and identify 224 integration hot spots. Investigation of the possible genome features involved in hot-spot formation revealed that the expression level of the genes did not influence distribution of the integration sites of gammaretroviruses (PERV and MLV) or the formation of integration hot spots. However, PERV integration and the presence of hot spots was found to be greater in areas of the genome with high densities of genes with CpG islands. Surprisingly, this was not true for MLV. Simulation of integration profiles revealed that retrovirus integration studies involving the use of the restriction enzyme MseI (widely used in genome integration studies of MLV and gammaretroviral vector) underestimated integration near CpG islands and in gene-dense areas. These results suggest that the integration of gammaretrovirus or gammaretroviral vectors might occur preferentially in genome areas that are highly enriched in genes under CpG island promoter regulation.
Collapse
|
172
|
Hacein-Bey-Abina S, Garrigue A, Wang GP, Soulier J, Lim A, Morillon E, Clappier E, Caccavelli L, Delabesse E, Beldjord K, Asnafi V, MacIntyre E, Dal Cortivo L, Radford I, Brousse N, Sigaux F, Moshous D, Hauer J, Borkhardt A, Belohradsky BH, Wintergerst U, Velez MC, Leiva L, Sorensen R, Wulffraat N, Blanche S, Bushman FD, Fischer A, Cavazzana-Calvo M. Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J Clin Invest 2008; 118:3132-42. [PMID: 18688285 DOI: 10.1172/jci35700] [Citation(s) in RCA: 1386] [Impact Index Per Article: 81.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2008] [Accepted: 06/25/2008] [Indexed: 12/16/2022] Open
Abstract
Previously, several individuals with X-linked SCID (SCID-X1) were treated by gene therapy to restore the missing IL-2 receptor gamma (IL2RG) gene to CD34+ BM precursor cells using gammaretroviral vectors. While 9 of 10 patients were successfully treated, 4 of the 9 developed T cell leukemia 31-68 months after gene therapy. In 2 of these cases, blast cells contained activating vector insertions near the LIM domain-only 2 (LMO2) proto-oncogene. Here, we report data on the 2 most recent adverse events, which occurred in patients 7 and 10. In patient 10, blast cells contained an integrated vector near LMO2 and a second integrated vector near the proto-oncogene BMI1. In patient 7, blast cells contained an integrated vector near a third proto-oncogene,CCND2. Additional genetic abnormalities in the patients' blast cells included chromosomal translocations, gain-of-function mutations activating NOTCH1, and copy number changes, including deletion of tumor suppressor gene CDKN2A, 6q interstitial losses, and SIL-TAL1 rearrangement. These findings functionally specify a genetic network that controls growth in T cell progenitors. Chemotherapy led to sustained remission in 3 of the 4 cases of T cell leukemia, but failed in the fourth. Successful chemotherapy was associated with restoration of polyclonal transduced T cell populations. As a result, the treated patients continued to benefit from therapeutic gene transfer.
Collapse
Affiliation(s)
- Salima Hacein-Bey-Abina
- Department of Biotherapy, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Université René Descartes, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
173
|
Howe SJ, Mansour MR, Schwarzwaelder K, Bartholomae C, Hubank M, Kempski H, Brugman MH, Pike-Overzet K, Chatters SJ, de Ridder D, Gilmour KC, Adams S, Thornhill SI, Parsley KL, Staal FJT, Gale RE, Linch DC, Bayford J, Brown L, Quaye M, Kinnon C, Ancliff P, Webb DK, Schmidt M, von Kalle C, Gaspar HB, Thrasher AJ. Insertional mutagenesis combined with acquired somatic mutations causes leukemogenesis following gene therapy of SCID-X1 patients. J Clin Invest 2008. [PMID: 18688286 DOI: 10.1172/jci35798ds1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
X-linked SCID (SCID-X1) is amenable to correction by gene therapy using conventional gammaretroviral vectors. Here, we describe the occurrence of clonal T cell acute lymphoblastic leukemia (T-ALL) promoted by insertional mutagenesis in a completed gene therapy trial of 10 SCID-X1 patients. Integration of the vector in an antisense orientation 35 kb upstream of the protooncogene LIM domain only 2 (LMO2) caused overexpression of LMO2 in the leukemic clone. However, leukemogenesis was likely precipitated by the acquisition of other genetic abnormalities unrelated to vector insertion, including a gain-of-function mutation in NOTCH1, deletion of the tumor suppressor gene locus cyclin-dependent kinase 2A (CDKN2A), and translocation of the TCR-beta region to the STIL-TAL1 locus. These findings highlight a general toxicity of endogenous gammaretroviral enhancer elements and also identify a combinatorial process during leukemic evolution that will be important for risk stratification and for future protocol design.
Collapse
Affiliation(s)
- Steven J Howe
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, University College London, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Staal FJT, Pike-Overzet K, Ng YY, van Dongen JJM. Sola dosis facit venenum. Leukemia in gene therapy trials: a question of vectors, inserts and dosage? Leukemia 2008; 22:1849-52. [PMID: 18769449 DOI: 10.1038/leu.2008.219] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2008] [Revised: 07/23/2008] [Accepted: 07/28/2008] [Indexed: 01/10/2023]
Abstract
In clinical gene therapy trials for X-linked severe combined immunodeficiency, the development of leukemia has come up as a severe adverse effect. In all five cases, T-cell acute lymphoblastic leukemia (T-ALL) occurred as a direct consequence of insertional mutagenesis by the retrovirus used to deliver the therapeutic gene. Here, we review the mechanisms of insertional mutagenesis, the function of the Il2RG gene and the future developments in the field. New lentiviral and gamma retroviral vectors can significantly improve the safety profile of the tools used but still carry the risk of insertional mutagenesis, as shown in this issue of Leukemia. Finally, the unfortunate side effects of gene therapy have given more insight into the development of human T-ALL.
Collapse
Affiliation(s)
- F J T Staal
- Department of Immunology, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | | | | | | |
Collapse
|
175
|
Moreno-Carranza B, Gentsch M, Stein S, Schambach A, Santilli G, Rudolf E, Ryser MF, Haria S, Thrasher AJ, Baum C, Brenner S, Grez M. Transgene optimization significantly improves SIN vector titers, gp91phox expression and reconstitution of superoxide production in X-CGD cells. Gene Ther 2008; 16:111-8. [PMID: 18784749 DOI: 10.1038/gt.2008.143] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Gene therapy has proven to be of potential value for the correction of inherited hematopoietic disorders. However, the occurrence of severe side effects in some of the clinical trials has questioned the safety of this approach and has hampered the use of long terminal repeat-driven vectors for the treatment of a large number of patients. The development of self-inactivating (SIN) vectors with reduced genotoxicity provides an alternative to the currently used vectors. Our initial attempts to use SIN vectors for the correction of a myeloid disorder, chronic granulomatous disease, failed due to low vector titers and poor transgene expression. The optimization of the transgene cDNA (gp91(phox)) resulted in substantially increased titers and transgene expression. Most notably, transgene optimization significantly improved expression of a second cistron located downstream of gp91(phox). Thus, optimization of the transgene sequence results in higher expression levels and increased therapeutic index allowing the use of low vector copy numbers per transduced cell and weaker internal promoters.
Collapse
Affiliation(s)
- B Moreno-Carranza
- Division of Applied Virology and Gene Therapy, Institute for Biomedical Research, Georg-Speyer-Haus, Frankfurt, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
176
|
Howe SJ, Mansour MR, Schwarzwaelder K, Bartholomae C, Hubank M, Kempski H, Brugman MH, Pike-Overzet K, Chatters SJ, de Ridder D, Gilmour KC, Adams S, Thornhill SI, Parsley KL, Staal FJ, Gale RE, Linch DC, Bayford J, Brown L, Quaye M, Kinnon C, Ancliff P, Webb DK, Schmidt M, von Kalle C, Gaspar HB, Thrasher AJ. Insertional mutagenesis combined with acquired somatic mutations causes leukemogenesis following gene therapy of SCID-X1 patients. J Clin Invest 2008; 118:3143-50. [PMID: 18688286 PMCID: PMC2496964 DOI: 10.1172/jci35798] [Citation(s) in RCA: 942] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Accepted: 06/25/2008] [Indexed: 12/27/2022] Open
Abstract
X-linked SCID (SCID-X1) is amenable to correction by gene therapy using conventional gammaretroviral vectors. Here, we describe the occurrence of clonal T cell acute lymphoblastic leukemia (T-ALL) promoted by insertional mutagenesis in a completed gene therapy trial of 10 SCID-X1 patients. Integration of the vector in an antisense orientation 35 kb upstream of the protooncogene LIM domain only 2 (LMO2) caused overexpression of LMO2 in the leukemic clone. However, leukemogenesis was likely precipitated by the acquisition of other genetic abnormalities unrelated to vector insertion, including a gain-of-function mutation in NOTCH1, deletion of the tumor suppressor gene locus cyclin-dependent kinase 2A (CDKN2A), and translocation of the TCR-beta region to the STIL-TAL1 locus. These findings highlight a general toxicity of endogenous gammaretroviral enhancer elements and also identify a combinatorial process during leukemic evolution that will be important for risk stratification and for future protocol design.
Collapse
Affiliation(s)
- Steven J. Howe
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Marc R. Mansour
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Kerstin Schwarzwaelder
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Cynthia Bartholomae
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Michael Hubank
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Helena Kempski
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Martijn H. Brugman
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Karin Pike-Overzet
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Stephen J. Chatters
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Dick de Ridder
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Kimberly C. Gilmour
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Stuart Adams
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Susannah I. Thornhill
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Kathryn L. Parsley
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Frank J.T. Staal
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Rosemary E. Gale
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - David C. Linch
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Jinhua Bayford
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Lucie Brown
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Michelle Quaye
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Christine Kinnon
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Philip Ancliff
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - David K. Webb
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Manfred Schmidt
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Christof von Kalle
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - H. Bobby Gaspar
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| | - Adrian J. Thrasher
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, and
Department of Haematology, University College London, London, United Kingdom.
Department of Translational Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, United Kingdom.
Paediatric Malignancy Cytogenetics Unit, Great Ormond Street Hospital, London, United Kingdom.
Department of Experimental Hematology, Hannover Medical School, Hannover, Germany.
Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands.
Information and Communication Theory Group, Faculty of Electrical Engineering, Mathematics, and Computer Science, Delft University of Technology, Delft, The Netherlands.
Great Ormond Street Hospital for Children NHS Trust, London, United Kingdom.
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, USA
| |
Collapse
|
177
|
Hu J, Renaud G, Golmes T, Ferris A, Hendrie PC, Donahue RE, Hughes SH, Wolfsberg TG, Russell DW, Dunbar CE. Reduced genotoxicity of avian sarcoma leukosis virus vectors in rhesus long-term repopulating cells compared to standard murine retrovirus vectors. Mol Ther 2008; 16:1617-23. [PMID: 18578011 PMCID: PMC2561952 DOI: 10.1038/mt.2008.135] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Insertional mutagenesis continues to be a major concern in hematopoietic stem-cell gene therapy. Nonconventional gene transfer vectors with more favorable integration features in comparison with conventional retrovirus and lentivirus vectors are being developed and optimized. In this study, we report for the first time a systematic analysis of 198 avian sarcoma leukosis virus (ASLV) insertion sites identified in rhesus long-term repopulating cells, and a comparison of ASLV insertion patterns to Moloney murine leukemia virus (MLV) (n = 396) and simian immunodeficiency virus (SIV) (n = 289) using the newly released rhesus genome databank. Despite a weak preference toward gene-coding regions, ASLV integration is nonclustered, does not favor gene-rich regions, transcription start sites, or CpG islands. There was no propensity for ASLV insertions within or near proto-oncogenes, and most importantly, no insertions close to or within the Mds1-Evi1 locus, which is in contrast to the significant over-representation of this insertion site for MLV vectors in the same transplantation model. Furthermore, ASLV long terminal repeats (LTRs) do not have detectable promoter and enhancer activity in a quantitative luciferase assay to measure neighboring gene activation. The combination of these features is unique for ASLV and suggests that optimized vectors based on this virus could be useful and safe for gene transfer to hematopoietic stem cells and progenitor cells.
Collapse
Affiliation(s)
- Jingqiong Hu
- Molecular Hematopoiesis Section, Hematology Branch, National Heart, Lung, Blood Institute, National Institute of Health, Bethesda, MD, 20892-1202
| | - Gabriel Renaud
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Theotonius Golmes
- Molecular Hematopoiesis Section, Hematology Branch, National Heart, Lung, Blood Institute, National Institute of Health, Bethesda, MD, 20892-1202
| | - Andrea Ferris
- HIV Drug Resistance Program National Cancer Institute at Frederick, Frederick, MD, 21702-1201
| | - Paul C. Hendrie
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA 98195, USA
| | - Robert E. Donahue
- Molecular Hematopoiesis Section, Hematology Branch, National Heart, Lung, Blood Institute, National Institute of Health, Bethesda, MD, 20892-1202
| | - Stephen H. Hughes
- HIV Drug Resistance Program National Cancer Institute at Frederick, Frederick, MD, 21702-1201
| | - Tyra G. Wolfsberg
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - David W. Russell
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA 98195, USA
| | - Cynthia E. Dunbar
- Molecular Hematopoiesis Section, Hematology Branch, National Heart, Lung, Blood Institute, National Institute of Health, Bethesda, MD, 20892-1202
| |
Collapse
|
178
|
Abstract
Although the remarkable versatility and efficacy of recombinant adeno-associated virus 2 (AAV2) vectors in transducing a wide variety of cells and tissues in vitro, and in numerous pre-clinical animal models of human diseases in vivo, have been well established, the published literature is replete with controversies with regard to the efficacy of AAV2 vectors in hematopoietic stem cell (HSC) transduction. A number of factors have contributed to these controversies, the molecular bases of which have begun to come to light in recent years. With the availability of several novel serotypes (AAV1 through AAV12), rational design of AAV capsid mutants, and strategies (self-complementary vector genomes, hematopoietic cell-specific promoters), it is indeed becoming feasible to achieve efficient transduction of HSC by AAV vectors. Using a murine serial bone marrow transplantation model in vivo, we have recently documented stable integration of the proviral AAV genome into mouse chromosomes, which does not lead to any overt hematological abnormalities. Thus, a better understanding of the AAV-HSC interactions, and the availability of a vast repertoire of novel serotype and capsid mutant vectors, are likely to have significant implications in the use of AAV vectors in high-efficiency transduction of HSCs as well as in gene therapy applications involving the hematopoietic system.
Collapse
Affiliation(s)
- Arun Srivastava
- Division of Cellular & Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida 32610-3633, USA.
| |
Collapse
|
179
|
Hacein-Bey-Abina S, Garrigue A, Wang GP, Soulier J, Lim A, Morillon E, Clappier E, Caccavelli L, Delabesse E, Beldjord K, Asnafi V, MacIntyre E, Dal Cortivo L, Radford I, Brousse N, Sigaux F, Moshous D, Hauer J, Borkhardt A, Belohradsky BH, Wintergerst U, Velez MC, Leiva L, Sorensen R, Wulffraat N, Blanche S, Bushman FD, Fischer A, Cavazzana-Calvo M. Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J Clin Invest 2008. [PMID: 18688285 DOI: 10.1172/jc135700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2023] Open
Abstract
Previously, several individuals with X-linked SCID (SCID-X1) were treated by gene therapy to restore the missing IL-2 receptor gamma (IL2RG) gene to CD34+ BM precursor cells using gammaretroviral vectors. While 9 of 10 patients were successfully treated, 4 of the 9 developed T cell leukemia 31-68 months after gene therapy. In 2 of these cases, blast cells contained activating vector insertions near the LIM domain-only 2 (LMO2) proto-oncogene. Here, we report data on the 2 most recent adverse events, which occurred in patients 7 and 10. In patient 10, blast cells contained an integrated vector near LMO2 and a second integrated vector near the proto-oncogene BMI1. In patient 7, blast cells contained an integrated vector near a third proto-oncogene,CCND2. Additional genetic abnormalities in the patients' blast cells included chromosomal translocations, gain-of-function mutations activating NOTCH1, and copy number changes, including deletion of tumor suppressor gene CDKN2A, 6q interstitial losses, and SIL-TAL1 rearrangement. These findings functionally specify a genetic network that controls growth in T cell progenitors. Chemotherapy led to sustained remission in 3 of the 4 cases of T cell leukemia, but failed in the fourth. Successful chemotherapy was associated with restoration of polyclonal transduced T cell populations. As a result, the treated patients continued to benefit from therapeutic gene transfer.
Collapse
Affiliation(s)
- Salima Hacein-Bey-Abina
- Department of Biotherapy, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Université René Descartes, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
180
|
Silver JN, Flotte TR. Towards a rAAV-based gene therapy for ADA-SCID: from ADA deficiency to current and future treatment strategies. Pharmacogenomics 2008; 9:947-68. [PMID: 18597656 DOI: 10.2217/14622416.9.7.947] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Adenosine deaminase deficiency fosters a rare, devastating pediatric immune deficiency with concomitant opportunistic infections, metabolic anomalies and multiple organ system pathology. The standard of care for adenosine deaminase deficient severe combined immune deficiency (ADA-SCID) includes enzyme replacement therapy or bone marrow transplantation. Gene therapies for ADA-SCID over nearly two decades have exclusively involved retroviral vectors targeted to lymphocytes and hematopoetic progenitors. These groundbreaking gene therapies represent a revolution in clinical medicine, but come with several challenges, including the risk of insertional mutagenesis. An alternative gene therapy for ADA-SCID may utilize recombinant adeno-associated virus vectors in vivo, with numerous target tissues, to foster ectopic expression and secretion of adenosine deaminase. This review endeavors to describe ADA-SCID, the traditional treatments, previous retroviral gene therapies, and primarily, alternative recombinant adeno-associated virus-based strategies to remedy this potentially fatal genetic disease.
Collapse
Affiliation(s)
- Jared N Silver
- University of Florida College of Medicine, Department of Pediatrics, Gainesville, FL 32607, USA.
| | | |
Collapse
|
181
|
Abstract
Primary immunodeficiencies are a group of disorders that are highly amenable to gene therapy because of their defined pathophysiology and the accessibility of the hematopoietic system to molecular intervention. The development of this new therapeutic modality has been driven by the established morbidity and mortality associated with conventional allogeneic stem cell transplantation, particularly in the human leukocyte antigen-mismatched setting. Recently, several clinical studies have shown that gamma retroviral gene transfer technology can produce major beneficial therapeutic effects, but, as for all cellular and pharmacologic treatment approaches, with a finite potential for toxicity. Newer developments in vector design showing promise in overcoming these issues are likely to establish gene therapy as an efficacious strategy for many forms of primary immunodeficiencies.
Collapse
Affiliation(s)
- Adrian J Thrasher
- Centre for Immunodeficiency, Molecular Immunology Unit, Institute of Child Health, University College London and Great Ormond Street Hospital for Children NHS Trust, 30 Guilford Street, London WC1N 1EH, UK.
| |
Collapse
|
182
|
En route to ethical recommendations for gene transfer clinical trials. Mol Ther 2008; 16:432-8. [PMID: 18297094 DOI: 10.1038/mt.2008.13] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
183
|
Daniel R, Smith JA. Integration site selection by retroviral vectors: molecular mechanism and clinical consequences. Hum Gene Ther 2008; 19:557-68. [PMID: 18533894 DOI: 10.1089/hum.2007.148] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Retroviral DNA integration into the host cell genome is an essential feature of the retroviral life cycle. The ability to integrate their DNA into the DNA of infected cells also makes retroviruses attractive vectors for delivery of therapeutic genes into the genome of cells carrying adverse mutations in their cellular DNA. Sequencing of the entire human genome has enabled identification of integration site preferences of both replication-competent retroviruses and retroviral vectors. These results, together with the unfortunate outcome of a gene therapy trial, in which integration of a retroviral vector in the vicinity of a protooncogene was associated with the development of leukemia, have stimulated efforts to elucidate the molecular mechanism underlying integration site selection by retroviral vectors, as well as the development of methods to direct integration to specific DNA sequences and chromosomal regions. This review outlines our current knowledge of the mechanism of integration site selection by retroviruses in vitro, in cultured cells, and in vivo; the outcome of several of the more recent gene therapy trials, which employed these vectors; and the efforts of several laboratories to develop vectors that integrate at predetermined sites in the human genome.
Collapse
Affiliation(s)
- René Daniel
- Division of Infectious Diseases, Center for Human Virology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | | |
Collapse
|
184
|
Abstract
The concept of introducing genes into human cells for therapeutic purposes developed nearly 50 years ago as diseases due to defects in specific genes were recognized. Development of recombinant DNA techniques in the 1970s and their application to the study of mouse tumor viruses facilitated the assembly of the first gene transfer vectors. Vectors of several different types have now been developed for specific applications and over the past decade, efficacy has been demonstrated in many animal models. Clinical trials began in 1989 and by 2002 there was unequivocal evidence that children with severe combined immunodeficiency could be cured by gene transfer into primitive hematopoietic cells. Emerging from these successful trials was the realization that proto-oncogene activation by retroviral integration could contribute to leukemia. Much current effort is focused on development of safer vectors. Successful gene therapy applications have also been developed for control of graft-versus-host disease and treatment of various viral infections, leukemias, and lymphomas. The hemophilias seem amenable to gene therapy intervention and informative clinical trials have been conducted. The hemoglobin disorders, an early target for gene therapy, have proved particularly challenging although ongoing research is yielding new information that may ultimately lead to successful clinical trials.
Collapse
|
185
|
Sokolic R, Kesserwan C, Candotti F. Recent advances in gene therapy for severe congenital immunodeficiency diseases. Curr Opin Hematol 2008; 15:375-80. [PMID: 18536577 PMCID: PMC2666972 DOI: 10.1097/moh.0b013e328302c807] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
PURPOSE OF REVIEW To discuss new data on the safety and efficacy of the ongoing gene therapy trials for primary immune deficiencies, the first reports of new trials and the preclinical developments that are likely to be translated to the clinic in the near future. RECENT FINDINGS Both clinical successes and severe adverse events continue to be reported in trials of gammaretroviral gene therapy for severe combined immune deficiency-X1, adenosine deaminase-deficient forms of severe combined immune deficiency and chronic granulomatous disease. Insertion site analyses of recently reported trials on all of these diseases have discovered preferential insertion in the 5' ends of genes, including potentially dangerous ones such as proto-oncogenes and signal transduction and proliferation genes. Preclinical work on rodent and canine models has tested novel vectors, including lentiviruses and foamy viruses. SUMMARY Gene therapy for the most common forms of severe combined immune deficiency can lead to immune reconstitution in most patients, although a minority of patients has derived minimal clinical benefit and some have suffered severe adverse events including death. Ongoing preclinical work attempts to address the latter shortcoming. Meanwhile, in the presence of a careful risk-benefit assessment, gene therapy remains an appropriate subject of clinical investigation.
Collapse
Affiliation(s)
- Robert Sokolic
- Staff Clinician, Disorders of Immunity Section, Genetics and Molecular Biology Branch, National Human Genome Research Institute, National Institutes of Health, 10 Center Drive, MSC 1451, Building 10-CRC, Room 6-3330, Bethesda, MD 20892-1451, Voice: 301-451-1498 FAX: 301-451-5408,
| | - Chimène Kesserwan
- Clinical Fellow, Disorders of Immunity Section, Genetics and Molecular Biology Branch, National Human Genome Research Institute, National Institutes of Health, 10 Center Drive, MSC 1611, Building 10-CRC, Room 6-3330, Bethesda, MD 20892-1611, Voice: 301-451-3949 FAX: 301-480-3015,
| | - Fabio Candotti
- Senior Investigator, Head, Disorders of Immunity Section, Genetics and Molecular Biology Branch, National Human Genome Research Institute, National Institutes of Health, 49 Convent Drive, MSC 4442, Building 49, Room 3A04, Bethesda, MD 20892-4442, Voice: 301-435-2944 FAX: 301-480-3678,
| |
Collapse
|
186
|
Abstract
The discovery of RNA interference (RNAi) has resulted in a new class of biological agents that can specifically downmodulate HIV-1 gene expression. Delivery of these RNAi-based agents and the emergence of viral resistance present pressing issues in the use of RNAi in a genetic-based therapy for HIV-1. Here, we discuss a potential avenue around viral resistance and a targeted delivery scheme for treating HIV-1-infected individuals involving transcriptional gene silencing. Specifically, the use of small antisense RNAs targeted to the viral promoter regions and delivery by lentiviral-based mobilization-competent vectors expressing these promoter targeted RNAs may prove therapeutically relevant in a genetic therapy-based approach to treating HIV-1 infection.
Collapse
Affiliation(s)
- Anne-Marie W Turner
- Department of Molecular & Experimental Medicine and Kellogg School of Science & Technology, The Scripps Research Institute, 10550 N. Torrey Pines Road, MEM-115, La Jolla, CA 92037, USA
| | - Kevin V Morris
- Department of Molecular & Experimental Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, MEM-115, La Jolla, CA 92037, USA
| |
Collapse
|
187
|
Maina N, Han Z, Li X, Hu Z, Zhong L, Bischof D, Weigel-Van Aken KA, Slayton WB, Yoder MC, Srivastava A. Recombinant self-complementary adeno-associated virus serotype vector-mediated hematopoietic stem cell transduction and lineage-restricted, long-term transgene expression in a murine serial bone marrow transplantation model. Hum Gene Ther 2008; 19:376-83. [PMID: 18370591 DOI: 10.1089/hum.2007.143] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Although conventional recombinant single-stranded adeno-associated virus serotype 2 (ssAAV2) vectors have been shown to efficiently transduce numerous cells and tissues such as brain and muscle, their ability to transduce primary hematopoietic stem cells (HSCs) has been reported to be controversial. We have previously documented that among the ssAAV serotype 1 through 5 vectors, ssAAV1 vectors are more efficient in transducing primary murine HSCs, but that viral second-strand DNA synthesis continues to be a rate-limiting step. In the present studies, we evaluated the transduction efficiency of several novel serotype vectors (AAV1, AAV7, AAV8, and AAV10) and documented efficient transduction of HSCs in a murine serial bone marrow transplantation model. Self-complementary AAV (scAAV) vectors were found to be more efficient than ssAAV vectors, and the use of hematopoietic cell-specific enhancers/promoters, such as the human beta-globin gene DNase I-hypersensitive site 2 enhancer and promoter (HS2-betap) from the beta-globin locus control region (LCR), and the human parvovirus B19 promoter at map unit 6 (B19p6), allowed sustained transgene expression in an erythroid lineage-restricted manner in both primary and secondary transplant recipient mice. The proviral AAV genomes were stably integrated into progenitor cell chromosomal DNA, and did not lead to any overt hematological abnormalities in mice. These studies demonstrate the feasibility of the use of novel scAAV vectors for achieving high-efficiency transduction of HSCs as well as erythroid lineage-restricted expression of a therapeutic gene for the potential gene therapy of beta-thalassemia and sickle cell disease.
Collapse
Affiliation(s)
- Njeri Maina
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
188
|
Hai M, Adler RL, Bauer TR, Tuschong LM, Gu YC, Wu X, Hickstein DD. Potential genotoxicity from integration sites in CLAD dogs treated successfully with gammaretroviral vector-mediated gene therapy. Gene Ther 2008; 15:1067-71. [PMID: 18369320 PMCID: PMC6291306 DOI: 10.1038/gt.2008.52] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Revised: 02/18/2008] [Accepted: 02/18/2008] [Indexed: 11/09/2022]
Abstract
Integration site analysis was performed on six dogs with canine leukocyte adhesion deficiency (CLAD) that survived greater than 1 year after infusion of autologous CD34+ bone marrow cells transduced with a gammaretroviral vector expressing canine CD18. A total of 387 retroviral insertion sites (RIS) were identified in the peripheral blood leukocytes from the six dogs at 1 year postinfusion. A total of 129 RIS were identified in CD3+ T-lymphocytes and 102 RIS in neutrophils from two dogs at 3 years postinfusion. RIS occurred preferentially within 30 kb of transcription start sites, including 40 near oncogenes and 52 near genes active in hematopoietic stem cells. Integrations clustered around common insertion sites more frequently than random. Despite potential genotoxicity from RIS, to date there has been no progression to oligoclonal hematopoiesis and no evidence that vector integration sites influenced cell survival or proliferation. Continued follow-up in disease-specific animal models such as CLAD will be required to provide an accurate estimate of the genotoxicity using gammaretroviral vectors for hematopoietic stem cell gene therapy.
Collapse
Affiliation(s)
- M Hai
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1203, USA
| | | | | | | | | | | | | |
Collapse
|
189
|
|
190
|
|
191
|
Modlich U, Schambach A, Brugman MH, Wicke DC, Knoess S, Li Z, Maetzig T, Rudolph C, Schlegelberger B, Baum C. Leukemia induction after a single retroviral vector insertion in Evi1 or Prdm16. Leukemia 2008; 22:1519-28. [PMID: 18496560 DOI: 10.1038/leu.2008.118] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Insertional activation of cellular proto-oncogenes by replication-defective retroviral vectors can trigger clonal dominance and leukemogenesis in animal models and clinical trials. Here, we addressed the leukemogenic potential of vectors expressing interleukin-2 receptor common gamma-chain (IL2RG), the coding sequence required for correction of X-linked severe combined immunodeficiency. Similar to conventional gamma-retroviral vectors, self-inactivating (SIN) vectors with strong internal enhancers also triggered profound clonal imbalance, yet with a characteristic insertion preference for a window located downstream of the transcriptional start site. Controls including lentivirally transduced cells revealed that ectopic IL2RG expression was not sufficient to trigger leukemia. After serial bone marrow transplantation involving 106 C57Bl6/J mice monitored for up to 18 months, we observed leukemic progression of six distinct clones harboring gamma-retroviral long terminal repeat (LTR) or SIN vector insertions in Evi1 or Prdm16, two functionally related genes. Three leukemic clones had single vector integrations, and identical clones manifested with a remarkably similar latency and phenotype in independent recipients. We conclude that upregulation of Evi1 or Prdm16 was sufficient to initiate a leukemogenic cascade with consistent intrinsic dynamics. Our study also shows that insertional mutagenesis is required for leukemia induction by IL2RG vectors, a risk to be addressed by improved vector design.
Collapse
Affiliation(s)
- U Modlich
- Department of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
192
|
Zhang XB, Beard BC, Trobridge GD, Wood BL, Sale GE, Sud R, Humphries RK, Kiem HP. High incidence of leukemia in large animals after stem cell gene therapy with a HOXB4-expressing retroviral vector. J Clin Invest 2008; 118:1502-10. [PMID: 18357342 DOI: 10.1172/jci34371] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Accepted: 02/06/2008] [Indexed: 12/15/2022] Open
Abstract
Retroviral vector-mediated HSC gene therapy has been used to treat individuals with a number of life-threatening diseases. However, some patients with SCID-X1 developed retroviral vector-mediated leukemia after treatment. The selective growth advantage of gene-modified cells in patients with SCID-X1 suggests that the transgene may have played a role in leukemogenesis. Here we report that 2 of 2 dogs and 1 of 2 macaques developed myeloid leukemia approximately 2 years after being transplanted with cells that overexpressed homeobox B4 (HOXB4) and cells transduced with a control gammaretroviral vector that did not express HOXB4. The leukemic cells had dysregulated expression of oncogenes, a block in myeloid differentiation, and overexpression of HOXB4. HOXB4 knockdown restored differentiation in leukemic cells, suggesting involvement of HOXB4. In contrast, leukemia did not arise from the cells carrying the control gammaretroviral vector. In addition, leukemia did not arise in 5 animals with high-level marking and polyclonal long-term repopulation following transplantation with cells transduced with an identical gammaretrovirus vector backbone expressing methylguanine methyltransferase. These findings, combined with the absence of leukemia in many other large animals transplanted with cells transduced with gammaretroviral vectors expressing genes other than HOXB4, show that HOXB4 overexpression poses a significant risk of leukemogenesis. Our data thus suggest the continued need for caution in genetic manipulation of repopulating cells, particularly when the transgene might impart an intrinsic growth advantage.
Collapse
Affiliation(s)
- Xiao-Bing Zhang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109-1024, USA
| | | | | | | | | | | | | | | |
Collapse
|
193
|
AAV-mediated gene transfer for the treatment of hemophilia B: problems and prospects. Gene Ther 2008; 15:870-5. [PMID: 18432276 DOI: 10.1038/gt.2008.71] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Adeno-associated viral vector-mediated gene transfer of coagulation factor IX to the skeletal muscle or to liver has resulted in sustained correction of hemophilia B in mice and dogs. The two initial phase I/II AAV clinical trials for hemophilia B, delivering a factor IX cDNA to skeletal muscle or liver, showed no serious adverse events. Although the muscle trial failed to achieve a therapeutic level of factor IX in the circulation, long-term expression of clotting factor was demonstrated on muscle biopsies taken up to 3 years after vector injection. Administration of vector to liver via the hepatic artery identified a therapeutic dose, which agreed closely with the doses predicted by studies in hemophilic dogs. However, expression in human subjects lasted for only a period of weeks, followed by a gradual decline in factor IX levels accompanied by a self-limited, asymptomatic rise and fall in liver enzymes. Immune responses to vector capsid may account for this difference in outcome between humans and other species. Here we review the results from both preclinical and clinical studies of adeno-associated viral vector gene transfer for hemophilia B, and the problems that have been identified and that must be overcome to achieve successful transduction and sustained expression.
Collapse
|
194
|
Santilli G, Thornhill SI, Kinnon C, Thrasher AJ. Gene therapy of inherited immunodeficiencies. Expert Opin Biol Ther 2008; 8:397-407. [PMID: 18352845 DOI: 10.1517/14712598.8.4.397] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Primary immunodeficiencies (PID) are a group of inherited diseases that affect the development or activity of the immune system. In severe cases allogeneic haematopoietic stem cell transplantation has proved to be a successful curative modality but it is limited by toxicity and reduced efficacy in mismatched donor settings. OBJECTIVE Gene therapy for PID has been developed as an alternative strategy and has entered the clinical arena. In this review we discuss the outcomes of recent gene therapy trials and some of the problems that remain to be tackled. METHODS Results from clinical trials for X-linked severe combined immunodeficiency (SCID-X1), adenosine deaminase deficient SCID (ADA-SCID), and X-linked chronic granulomatous disease (X-CGD) are discussed. In addition, other conditions are highlighted such as the Wiskott Aldrich Syndrome (WAS) for which gene therapy has shown considerable promise in preclinical studies, and are currently being translated into novel clinical approaches. RESULTS/CONCLUSION Whilst these encouraging results demonstrate that gene therapy can be used successfully to treat monogenic PID, the occurrence of vector-related side effects has highlighted the need for accurate assessment of the associated risks and a requirement for improvements in vector design.
Collapse
Affiliation(s)
- Giorgia Santilli
- University College London, Institute of Child Health, Centre for Immunodeficiency, Molecular Immunology Unit, 30 Guilford Street, London, WC1N 1EH, UK
| | | | | | | |
Collapse
|
195
|
Absence of retroviral vector-mediated transformation of gene-modified T cells after long-term engraftment in mice. Gene Ther 2008; 15:1056-66. [DOI: 10.1038/gt.2008.47] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
196
|
Thornhill SI, Schambach A, Howe SJ, Ulaganathan M, Grassman E, Williams D, Schiedlmeier B, Sebire NJ, Gaspar HB, Kinnon C, Baum C, Thrasher AJ. Self-inactivating gammaretroviral vectors for gene therapy of X-linked severe combined immunodeficiency. Mol Ther 2008; 16:590-8. [PMID: 18180772 PMCID: PMC6748866 DOI: 10.1038/sj.mt.6300393] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Gene therapy for X-linked severe combined immunodeficiency (SCID-X1) has proven highly effective for long-term restoration of immunity in human subjects. However, the development of lymphoproliferative complications due to dysregulated proto-oncogene expression has underlined the necessity for developing safer vector systems. To reduce the potential for insertional mutagenesis, we have evaluated the efficacy of self-inactivating (SIN) gammaretroviral vectors in cellular and in vivo models of SCID-X1. Vectors incorporating an internal human elongation factor-1alpha regulatory element were capable of fully restoring the lymphoid differentiation potential of gammac-deficient lineage negative cells. Multilineage lymphoid reconstitution of a murine model was achieved at a similar level to that achieved by a conventional long-terminal repeat (LTR)-regulated vector used in previous clinical trials. Functional proliferative responses to mitogenic stimuli were also restored, and serum immunoglobulin levels were normalized. The reduced mutagenic potential conferred by SIN vector configurations and alternative non-LTR-based regulatory elements, together with proven efficacy in correction of cellular defects provides an important platform for development of the next phase of clinical trials for SCID-X1.
Collapse
Affiliation(s)
- Susannah I Thornhill
- Centre for Immunodeficiency, Molecular Immunology Unit, Institute of Child Health, University College London, London, UK
| | - Axel Schambach
- Department of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Steven J Howe
- Centre for Immunodeficiency, Molecular Immunology Unit, Institute of Child Health, University College London, London, UK
| | - Meera Ulaganathan
- Centre for Immunodeficiency, Molecular Immunology Unit, Institute of Child Health, University College London, London, UK
| | - Elke Grassman
- Division of Experimental Hematology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - David Williams
- Division of Experimental Hematology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | | | - Neil J Sebire
- Department of Histopathology, Great Ormond Street Children’s Hospital, London, UK
| | - H Bobby Gaspar
- Centre for Immunodeficiency, Molecular Immunology Unit, Institute of Child Health, University College London, London, UK
- Department of Clinical Immunology, Great Ormond Street Hospital, London, UK
| | - Christine Kinnon
- Centre for Immunodeficiency, Molecular Immunology Unit, Institute of Child Health, University College London, London, UK
| | - Christopher Baum
- Department of Experimental Hematology, Hannover Medical School, Hannover, Germany
- Division of Experimental Hematology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Adrian J Thrasher
- Centre for Immunodeficiency, Molecular Immunology Unit, Institute of Child Health, University College London, London, UK
- Department of Clinical Immunology, Great Ormond Street Hospital, London, UK
| |
Collapse
|
197
|
Han Z, Zhong L, Maina N, Hu Z, Li X, Chouthai NS, Bischof D, Weigel-Van Aken KA, Slayton WB, Yoder MC, Srivastava A. Stable Integration of Recombinant Adeno-Associated Virus Vector Genomes After Transduction of Murine Hematopoietic Stem Cells. Hum Gene Ther 2008; 19:267-78. [DOI: 10.1089/hum.2007.161] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Zongchao Han
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610
| | - Li Zhong
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610
- Shands Cancer Center, University of Florida College of Medicine, Gainesville, FL 32610
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32610
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32610
| | - Njeri Maina
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Zhongbo Hu
- Division of Hematology/Oncology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610
| | - Xiaomiao Li
- Division of Hematology/Oncology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610
| | - Nitin S. Chouthai
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610
- Department of Pediatrics, Wayne State University, Detroit, MI 48201
| | - Daniela Bischof
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Kirsten A. Weigel-Van Aken
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610
- Shands Cancer Center, University of Florida College of Medicine, Gainesville, FL 32610
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32610
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32610
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL 32610
| | - William B. Slayton
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32610
- Division of Hematology/Oncology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610
| | - Mervin C. Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Arun Srivastava
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610
- Shands Cancer Center, University of Florida College of Medicine, Gainesville, FL 32610
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32610
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32610
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL 32610
| |
Collapse
|
198
|
Efficient protein expression from the endogenous RNA polymerase I promoter using a human ribosomal DNA targeting vector. Biochem Biophys Res Commun 2008; 367:846-51. [DOI: 10.1016/j.bbrc.2008.01.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2007] [Accepted: 01/03/2008] [Indexed: 12/17/2022]
|
199
|
Baum C, Schambach A, Modlich U, Thrasher A. [Gene therapy of SCID-X1]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2008; 50:1507-17. [PMID: 18046520 DOI: 10.1007/s00103-007-0385-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
X-linked severe combined immunodeficiency (SCID-X1) is an inherited disease caused by inactivating mutations in the gene encoding the interleukin 2 receptor common gamma chain (IL2RG), which is located on the X-chromosome. Affected boys fail to develop two major effector cell types of the immune system (T cells and NK cells) and suffer from a functional B cell defect. Although drugs such as antibiotics can offer partial protection, the boys normally die in the first year of life in the absence of a curative therapy. For a third of the children, bone marrow transplantation from a fully matched donor is available and can cure the disease without major side effects. Mismatched bone marrow transplantation, however, is complicated by severe and potentially lethal side effects. Over the past decade, scientists worldwide have developed new treatments by introducing a correct copy of the IL2RG-cDNA. Gene therapy was highly effective when applied in young children. However, in a few patients the IL2RG-gene vector has unfortunately caused leukaemia. Activation of cellular proto-oncogenes by accidental integration of the gene vector has been identified as the underlying mechanism. In future clinical trials, improved vector technology in combination with other protocol modifications may reduce the risk of this side effect.
Collapse
Affiliation(s)
- C Baum
- Medizinische Hochschule Hannover, BRD.
| | | | | | | |
Collapse
|
200
|
Globin lentiviral vector insertions can perturb the expression of endogenous genes in beta-thalassemic hematopoietic cells. Mol Ther 2008; 16:525-33. [PMID: 18195719 DOI: 10.1038/sj.mt.6300394] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Although hematopoietic cell gene therapy using retroviral vectors has recently achieved success in clinical trials, safety issues regarding vector insertional mutagenesis have emerged. Vector insertion, resulting in transcriptional activation of proto-oncogenes, played a role in the development of lymphoid leukemia in an X-linked severe combined immunodeficiency trial, and caused myeloid clonal dominance in a trial for chronic granulomatous disease. These events have raised the question of whether gene therapy for other disorders such as beta-thalassemia and sickle cell disease may hold a similar risk. In this study, we prospectively evaluated whether gamma-globin lentiviral vectors containing enhancer elements from the beta-globin locus control region could alter the expression of genes near the vector insertion. We studied this question in primary, clonal murine beta-thalassemic erythroid cells, where globin regulatory elements are highly active. We found an overall incidence of perturbed expression in 28% of the transduced clones, with 11% of all genes contained within a 600-kilobase region surrounding the vector-insertion site demonstrating altered expression. This rate was higher than that observed for a lentiviral vector containing a viral long-terminal repeat (LTR). This is the first direct evidence that lentiviral vectors can cause insertional dysregulation of cellular genes at a frequent rate.
Collapse
|