51
|
Abstract
Gene therapy is a very attractive strategy in experimental cancer therapy. Ideally, the approach aims to deliver therapeutic genes selectively to cancer cells. However, progress in the improvement of gene therapy formulations has been hampered by difficulties in measuring transgene delivery and in quantifying transgene expression in vivo. In clinical trials, endpoints rely almost exclusively on the analysis of biopsies, which provide limited information. Non-invasive monitoring of gene delivery and expression is a very attractive approach as it can be repeated over time in the same patient to provide spatiotemporal information on gene expression on a whole body scale. Thus, imaging methods can uniquely provide researchers and clinicians the ability to directly and serially assess morphological, functional and metabolic changes consequent to molecular and cellular based therapies. This review highlights the various methods currently being developed in preclinical models.
Collapse
|
52
|
Swainson L, Mongellaz C, Adjali O, Vicente R, Taylor N. Lentiviral Transduction of Immune Cells. Innate Immun 2008; 415:301-20. [DOI: 10.1007/978-1-59745-570-1_18] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/12/2023] Open
|
53
|
Abstract
This chapter describes the methods we use to transduce mouse and human hematopoietic stem cells (HSCs) and human embryonic stem cells (hESCs). We provide detailed protocols for producing high-titer lentiviral supernatants by transient transfection and for measuring viral titers. Methods to concentrate viral supernatants to achieve a higher titer are also described. The protocols given here have been used successfully to transduce engrafting mouse and human HSCs as well as progenitor cells. These cells maintained stable transgene expression after engraftment in mice and in vivo differentiation. Human ESCs can also be transduced with a high efficiency, and transgene is expressed stably after hematopoietic differentiation.
Collapse
Affiliation(s)
- Zhaohui Ye
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | |
Collapse
|
54
|
Abstract
Active cancer immunotherapy relies on functional tumor-specific effector T lymphocytes for tumor elimination. Dendritic cells (DCs), as most potent antigen-presenting cells, have been popularly employed in clinical and experimental tumor treatments. We have previously demonstrated that lentiviral vector-mediated transgene delivery to DC progenitors, including bone marrow cells and hematopoietic stem cells, followed by transplantation supports systemic generation of great numbers of tumor antigen-presenting DCs. These DCs subsequently stimulate marked and systemic immune activation. Here, we examined whether this level of immune activation is sufficient to overcome tumor-induced tolerogenic environment for treating an established aggressive epithelial tumor. We showed that a combination treatment of granulocyte macrophage-colony stimulating factor and cytosine-phosphate-guanine-containing oligonucleotide stimulated large numbers of tumor antigen-presenting DCs in situ from transgene-modified stem cells. Moreover, these in situ generated and activated DCs markedly stimulated activation of antigen-specific CD4 and CD8 T cells by augmenting their numbers, as well as function, even in a tumor-bearing tolerogenic environment. This leads to significant improvement in the therapeutic efficacy of established pulmonary metastases. This study suggests that lentiviral vector-modified stem cells as DC progenitors may be used as an effective therapeutic regimen for treating metastatic epithelial tumors.
Collapse
|
55
|
Breckpot K, Thielemans K. Lentiviruses in cancer immunotherapy. Future Virol 2007. [DOI: 10.2217/17460794.2.6.597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/21/2022]
Abstract
Lentiviral vectors have emerged as promising tools for cancer immunotherapy owing to their capacity to transduce a wide range of different cell types, including dendritic cells (DCs), the key regulators of immunity. Ex vivo transduced DCs proved to be potent inducers of strong antigen-specific T-cell responses, both in vitro and in vivo. Moreover, lentiviral vectors have been successfully applied for antigen-specific immunization, offering the advantage that the same lentivirus can be used for all patients resulting in an ‘off-the-shelf’ therapeutic. This review provides an update on the state-of-the-art induction of tumor-specific immune responses in vivo upon direct administration of tumor-associated antigen-encoding lentiviruses. Focusing on the cell types transduced, the results of current studies and the explanation for the potency of lentiviral vectors are discussed.
Collapse
Affiliation(s)
- Karine Breckpot
- Medical School of the Vrije Universiteit Brussel, Laboratory of Molecular & Cellular Therapy, Department of Physiology-Immunology, Laarbeeklaan 103 Building E, B-1090, Brussels, Belgium
| | - K Thielemans
- Medical School of the Vrije Universiteit Brussel, Laboratory of Molecular & Cellular Therapy, Department of Physiology-Immunology, Laarbeeklaan 103 Building E, B-1090, Brussels, Belgium
| |
Collapse
|
56
|
Lü P, Liu F, Yan L, Peng T, Liu T, Yao Z, Wang CY. Stem cells therapy for type 1 diabetes. Diabetes Res Clin Pract 2007; 78:1-7. [PMID: 17349714 DOI: 10.1016/j.diabres.2007.02.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 08/28/2006] [Revised: 01/10/2007] [Accepted: 02/02/2007] [Indexed: 11/30/2022]
Abstract
In this article, we have reviewed the developments of studies of stem cells therapy for type 1 diabetes since this century. Review of the literature was based on computer searches (PubMed) and our studies. Type 1 diabetes can now be ameliorated by islet transplantation, but this treatment is restricted by the scarcity of islet tissue. Hopes for a limitless supply of a substitute for primary islets of Langerhans and progress in stem cell biology have led to research into the feasibility of stem/progenitor cells to generate insulin-producing cells to use in replacement therapies for diabetes. An increasing body of evidence indicated that, in addition to embryonic stem cells, several potential adult stem/progenitor cells, derived from pancreas, liver, spleen, and bone marrow could differentiate into insulin-producing cells in vitro or in vivo. However, significant controversy currently exists in this field. Moreover, safe suppression of autoimmunity or specific tolerance to auto-antigens for patients with type 1 diabetes must be achieved before this promising new technology can lead to a great progress in clinical practice. To prevent type 1 diabetes through genetic engineering of hematopoietic stem cells represents another new strategy. Much basic research is still required.
Collapse
Affiliation(s)
- Ping Lü
- Department of General Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | | | | | | | | | | | | |
Collapse
|
57
|
Laurie KL, Blundell MP, Baxendale HE, Howe SJ, Sinclair J, Qasim W, Brunsberg U, Thrasher AJ, Holmdahl R, Gustafsson K. Cell-specific and efficient expression in mouse and human B cells by a novel hybrid immunoglobulin promoter in a lentiviral vector. Gene Ther 2007; 14:1623-31. [PMID: 17851547 DOI: 10.1038/sj.gt.3303021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/03/2023]
Abstract
The expression of genes specifically in B cells is of great interest in both experimental immunology as well as in future clinical gene therapy. We have constructed a novel enhanced B cell-specific promoter (Igk-E) consisting of an immunoglobulin kappa (Igk) minimal promoter combined with an intronic enhancer sequence and a 3' enhancer sequence from Ig genes. The Igk-E promoter was cloned into a lentiviral vector and used to control expression of enhanced green fluorescent protein (eGFP). Transduction of murine B-cell lymphoma cell lines and activated primary splenic B cells, with IgK-E-eGFP lentivirus, resulted in expression of eGFP, as analysed by flow cytometry, whereas expression in non-B cells was absent. The specificity of the promoter was further examined by transducing Lin(-) bone marrow with Igk-E-eGFP lentivirus and reconstituting lethally irradiated mice. After 16 weeks flow cytometry of lymphoid tissues revealed eGFP expression by CD19+ cells, but not by CD3+, CD11b+, CD11c+ or Gr-1+ cells. CD19+ cells were comprised of both marginal zone B cells and recirculating follicular B cells. Activated human peripheral mononuclear cells were also transduced with Igk-E-eGFP lentivirus under conditions of selective B-cell activation. The Igk-E promoter was able to drive expression of eGFP only in CD19+ cells, while eGFP was expressed by both spleen focus-forming virus and cytomegalovirus constitutive promoters in CD19+ and CD3+ lymphocytes. These data demonstrate that in these conditions the Igk-E promoter is cell specific and controls efficient expression of a reporter protein in mouse and human B cells in the context of a lentiviral vector.
Collapse
Affiliation(s)
- K L Laurie
- Molecular Immunology Unit, Wolfson Centre for Gene Therapy of Childhood Disease, UCL Institute of Child Health, London, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Cherqui S, Kingdon KM, Thorpe C, Kurian SM, Salomon DR. Lentiviral Gene Delivery of vMIP-II to Transplanted Endothelial Cells and Endothelial Progenitors Is Proangiogenic In Vivo. Mol Ther 2007; 15:1264-72. [PMID: 17505479 DOI: 10.1038/sj.mt.6300183] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/26/2022] Open
Abstract
Therapies that stimulate angiogenesis show promise in revascularization of transplanted or ischemic tissues. Viral macrophage inflammatory protein-II (vMIP-II) is encoded by human herpesvirus 8, and it can be both immunosuppressive and proangiogenic. However, little has been done to characterize the potential of vMIP-II-induced angiogenesis. We engineered a vMIP-II lentiviral gene vector, transduced both mature endothelial cells and progenitors, and transplanted these in Matrigel templates as an in vivo angiogenesis model. Our results show that vMIP-II promotes new, functional, branching, and segmented vessels associated with smooth muscle cells and connected with the host vasculature. Angiogenesis is enhanced through host cells as well as through transplanted vMIP-expressing endothelial cells. As a proof-of-concept for using vMIP-II in clinical applications, we showed that islets co-transplanted with endothelial cells expressing vMIP-II were revascularized and survived in Matrigel templates, whereas no islets survived under control conditions. vMIP-II up-regulates the expression of multiple proangiogenic factors that can have a synergistic effect. These include vascular endothelial growth factor (VEGF), kinase insert domain receptor, neuropilin 2, carcinoembryonic antigen-related cell adhesion molecule 1, interleukin-1alpha, fibronectin, and integrins alpha3, alpha4, and alpha5. These results provide the first demonstration that vMIP-II is proangiogenic in vivo and can deliver this function to endothelial progenitors as well as to mature endothelial cells through vector-mediated gene delivery.
Collapse
Affiliation(s)
- Stephanie Cherqui
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California, USA
| | | | | | | | | |
Collapse
|
59
|
Kimura T, Koya RC, Anselmi L, Sternini C, Wang HJ, Comin-Anduix B, Prins RM, Faure-Kumar E, Rozengurt N, Cui Y, Kasahara N, Stripecke R. Lentiviral Vectors with CMV or MHCII Promoters Administered In Vivo: Immune Reactivity Versus Persistence of Expression. Mol Ther 2007; 15:1390-9. [PMID: 17505480 DOI: 10.1038/sj.mt.6300180] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/09/2022] Open
Abstract
Lentiviral vectors (LVs) are potential tools for genetic vaccination. To improve the safety of LV vaccines, we evaluated the selectivity, bio-distribution, persistence of expression, and immune potency of vesicular stomatitis virus G (VSV-G)-pseudotyped vectors transcriptionally targeted to antigen presenting cells (APCs) through a major histocompatibility complex class II (MHCII) promoter. Control vectors contained the ubiquitous cytomegalovirus (CMV) promoter. Bio-distribution studies after intravenous injections of LVs expressing green fluorescent protein (GFP) or luciferase were conducted by a combination of flow cytometry, immunofluorescence, real-time quantitative polymerase chain reaction (RT-Q-PCR) and whole-body bioluminescence analyses. GFP-expressing vectors showed selective expression in MHCII(+) cells of spleen and LV-CMV-GFP administration produced noticeable spleen inflammation, whereas LV-MHCII-GFP did not. Long-term optical imaging analyses of C57BL/6 mice injected with LV-CMV-LUC showed diminishing luciferase expression in the liver and spleen over time. Vaccination/boost with LV-CMV expressing the melanoma antigen tyrosinase-related protein 2 (TRP2) yielded dose-dependent antigen-specific CD8(+) T-cell reactivity and high protection against B16 melanoma challenge. Unexpectedly, administration of LVs containing the MHCII promoter resulted in persistence of luciferase expression and viral integration in MHCII(+) splenocytes and virtually no CD8(+) T-cell responses against TRP2. These studies reveal that APC transduction by LVs could lead to immune reactivity (LV-CMV) or persistence of transgene expression (LV-MHCII), providing a relevant paradigm for vaccination and gene replacement approaches.
Collapse
Affiliation(s)
- Takahiro Kimura
- Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Breckpot K, Aerts JL, Thielemans K. Lentiviral vectors for cancer immunotherapy: transforming infectious particles into therapeutics. Gene Ther 2007; 14:847-62. [PMID: 17361214 DOI: 10.1038/sj.gt.3302947] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/12/2022]
Abstract
Lentiviral vectors have emerged as promising tools for both gene therapy and immunotherapy purposes. They exhibit several advantages over other viral systems in that they are less immunogenic and are capable of transducing a wide range of different cell types, including dendritic cells (DC). DC transduced ex vivo with a whole range of different (tumor) antigens were capable of inducing strong antigen-specific T-cell responses, both in vitro and in vivo. Recently, the administration of lentiviral vectors in vivo has gained substantial interest as an alternative method for antigen-specific immunization. This method offers a number of advantages over DC vaccines as the same lentivirus can in principle be used for all patients resulting in a significantly reduced cost and requirement for considerably less expertise for the generation and administration of lentiviral vaccines. By selectively targeting lentiviral vectors to, or restricting transgene expression in certain cell types, selectivity, safety and efficacy can be further improved. This review will focus on the use of direct administration of lentiviral vectors encoding tumor-associated antigens (TAA) for the induction of tumor-specific immune responses in vivo, with a special focus on problems related to the generation of large amounts of highly purified virus and specific targeting of antigen-presenting cells (APC).
Collapse
Affiliation(s)
- K Breckpot
- Laboratory of Molecular and Cellular Therapy, Department of Physiology and Immunology, Medical School of the Vrije Universiteit Brussel, Brussels, Belgium.
| | | | | |
Collapse
|
61
|
Chang AH, Sadelain M. The Genetic Engineering of Hematopoietic Stem Cells: the Rise of Lentiviral Vectors, the Conundrum of the LTR, and the Promise of Lineage-restricted Vectors. Mol Ther 2007; 15:445-56. [PMID: 17228317 DOI: 10.1038/sj.mt.6300060] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/20/2023] Open
Abstract
Recent studies on the integration patterns of different categories of retroviral vectors, the genotoxicity of long-terminal repeats (LTRs) and other genetic elements, the rise of lentiviral technology and the emergence of regulated vector systems providing tissue-restricted transgene expression and RNA interference, are profoundly changing the landscape of stem cell-based therapies. New developments in vector design and an increasing understanding of the mechanisms underlying insertional oncogenesis are ushering in a new phase in hematopoietic stem cell (HSC) engineering, thus bringing the hitherto exclusive reliance on LTR-driven, gamma-retroviral vectors to an end. Based on their ability to transduce non-dividing cells and their genomic stability, lentiviral vectors offer new prospects for the manipulation of HSCs. Tissue-specific vectors, as exemplified by globin vectors, not only provide therapeutic efficacy, but may also enhance safety, insofar that they restrict transgene expression in stem cells, progenitor cells and blood cells in all but the transcriptionally targeted lineage. This review provides a survey of these advances as well as several remaining challenges, focusing in particular on the importance of achieving adequate levels of protein expression from a limited number of vector copies per cell-ideally one to two.
Collapse
Affiliation(s)
- Alex H Chang
- Laboratory of Gene Transfer and Gene Expression, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | |
Collapse
|
62
|
Peacock CD, Wang Q, Gesell GS, Corcoran-Schwartz IM, Jones E, Kim J, Devereux WL, Rhodes JT, Huff CA, Beachy PA, Watkins DN, Matsui W. Hedgehog signaling maintains a tumor stem cell compartment in multiple myeloma. Proc Natl Acad Sci U S A 2007; 104:4048-53. [PMID: 17360475 PMCID: PMC1805487 DOI: 10.1073/pnas.0611682104] [Citation(s) in RCA: 344] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/21/2022] Open
Abstract
The cancer stem cell hypothesis suggests that malignant growth depends on a subset of tumor cells with stem cell-like properties of self-renewal. Because hedgehog (Hh) signaling regulates progenitor cell fate in normal development and homeostasis, aberrant pathway activation might be involved in the maintenance of such a population in cancer. Indeed, mutational activation of the Hh pathway is associated with medulloblastoma and basal cell carcinoma; pathway activity is also critical for growth of other tumors lacking such mutations, although the mechanism of pathway activation is poorly understood. Here we study the role and mechanism of Hh pathway activation in multiple myeloma (MM), a malignancy with a well defined stem cell compartment. In this model, rare malignant progenitors capable of clonal expansion resemble B cells, whereas the much larger tumor cell population manifests a differentiated plasma cell phenotype that pathologically defines the disease. We show that the subset of MM cells that manifests Hh pathway activity is markedly concentrated within the tumor stem cell compartment. The Hh ligand promotes expansion of MM stem cells without differentiation, whereas the Hh pathway blockade, while having little or no effect on malignant plasma cell growth, markedly inhibits clonal expansion accompanied by terminal differentiation of purified MM stem cells. These data reveal that Hh pathway activation is heterogeneous across the spectrum of MM tumor stem cells and their more differentiated progeny. The potential existence of similar relationships in other adult cancers may have important biologic and clinical implications for the study of aberrant Hh signaling.
Collapse
Affiliation(s)
| | - Qiuju Wang
- *Sidney Kimmel Comprehensive Cancer Center and
| | | | | | - Evan Jones
- *Sidney Kimmel Comprehensive Cancer Center and
| | - Jynho Kim
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21231; and
- Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305
| | | | | | | | - Philip A. Beachy
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21231; and
- Department of Developmental Biology, Institute for Stem Cell Biology and Regenerative Medicine, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305
- To whom correspondence may be addressed. E-mail:
, , or
| | - D. Neil Watkins
- *Sidney Kimmel Comprehensive Cancer Center and
- To whom correspondence may be addressed. E-mail:
, , or
| | - William Matsui
- *Sidney Kimmel Comprehensive Cancer Center and
- To whom correspondence may be addressed. E-mail:
, , or
| |
Collapse
|
63
|
Zhou BY, Ye Z, Chen G, Gao ZP, Zhang YA, Cheng L. Inducible and Reversible Transgene Expression in Human Stem Cells After Efficient and Stable Gene Transfer. Stem Cells 2006; 25:779-89. [PMID: 17158240 DOI: 10.1634/stemcells.2006-0128] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/17/2022]
Abstract
We report here a lentiviral vector system for regulated transgene expression. We used the tetracycline repressor fused with a transcriptional suppression domain (tTS) to specifically suppress transgene expression. Human cells were first transduced with a tTS-expressing vector and subsequently transduced with a second lentiviral vector-containing transgene controlled by a regular promoter adjacent to a high-affinity tTS-binding site (tetO). After optimizing the location of the tetO site in the latter vector, we achieved a better inducible transgene expression than the previous lentiviral vectors using the tetracycline repressor systems. In this new system, the transgene transcription from a cellular promoter such as EF1alpha or ubiquitin-C promoter is suppressed by the tTS bound to the nearby tetO site. In the presence of the tetracycline analog doxycycline (Dox), however, the tTS binding is released from the transgene vector and transcription from the promoter is restored. Thus, this system simply adds an extra level of regulation, suitable for any types of promoters (ubiquitous or cell-specific). We tested this tTS-suppressive, Dox-inducible system in 293T cells, human multipotent hematopoietic progenitor cells, and three human embryonic stem cell lines, using a dual-gene vector containing the green fluorescent protein reporter or a cellular gene. We observed a tight suppression in the uninduced state. However, the suppression is reversible, and transgene expression was restored at 5 ng/ml Dox. The lentiviral vectors containing the tTS-suppressive, Dox-inducible system offer a universal, inducible, and reversible transgene expression system in essentially any mammalian cell types, including human embryonic stem cells.
Collapse
Affiliation(s)
- Betty Ying Zhou
- Stem Cell Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Broadway Research Building, Room 747, 733 North Broadway, Baltimore, MD 21205, USA
| | | | | | | | | | | |
Collapse
|
64
|
Zhao P, Liu W, Cui Y. Rapid immune reconstitution and dendritic cell engraftment post–bone marrow transplantation with heterogeneous progenitors and GM-CSF treatment. Exp Hematol 2006; 34:951-64. [PMID: 16797423 DOI: 10.1016/j.exphem.2006.04.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/11/2006] [Revised: 03/21/2006] [Accepted: 04/10/2006] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Bone marrow/hematopoietic stem cell transplantation (BMT) has been the treatment of choice for severe hematological diseases and cancers. Rapid host immune recovery following BMT is critical for reducing complications and improving therapeutic outcome. Here we report manipulations that facilitate rapid immune and dendritic cell (DC) reconstitution post-BMT for improvement in therapeutic outcome of BMT-based disease treatment. METHODS Using lentiviral vector-modified or unmodified murine hematopoietic stem cells, we examined the engraftment efficiency and kinetics in immune reconstitution of unfractionated bone marrow cells (BM), lineage marker-negative (Lin-) hematopoietic progenitor cells (HPC), or purified Lin-Sca-1+ hematopoietic stem cells (HSC) at an equal hematopoietic progenitor number. RESULTS Our study revealed that BM reconstituted host primary and secondary lymphoid tissues more efficiently and rapidly. Moreover, in a competitive BMT setting using lentiviral vector-engineered BM and HSC expressing GFP or DsRed respectively, we showed that GM-CSF treatment further enhanced DC reconstitution to therapeutic relevant level as early as 2 weeks post-BMT. On the other hand, Flt3 ligand was less effective in enhancing DC reconstitution till 3 weeks post-BMT. This accelerated DC engraftment by GM-CSF treatment correlated well with improved overall immune reconstitution and enhanced activation of antigen-specific T cells post-BMT. CONCLUSION This study suggests that use of heterogeneous BM for transplantation facilitates more rapid immune reconstitution, especially in the presence of DC-stimulating cytokines. This improved immune reconstitution would provide additional therapeutic benefits for BMT-based immunotherapy and gene therapy of genetic disorders and cancers.
Collapse
Affiliation(s)
- Peilin Zhao
- Gene Therapy Program, Departments of Medicine and Genetics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | | | | |
Collapse
|
65
|
Dan YY, Riehle KJ, Lazaro C, Teoh N, Haque J, Campbell JS, Fausto N. Isolation of multipotent progenitor cells from human fetal liver capable of differentiating into liver and mesenchymal lineages. Proc Natl Acad Sci U S A 2006; 103:9912-7. [PMID: 16782807 PMCID: PMC1502553 DOI: 10.1073/pnas.0603824103] [Citation(s) in RCA: 234] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/12/2022] Open
Abstract
Little is known about the differentiation capabilities of nonhematopoietic cells of the human fetal liver. We report the isolation and characterization of a human fetal liver multipotent progenitor cell (hFLMPC) population capable of differentiating into liver and mesenchymal cell lineages. Human fetal livers (74-108 days of gestation) were dissociated and maintained in culture. We treated the colonies with geneticin and mechanically isolated hFLMPCs, which were kept in an undifferentiated state by culturing on feeder layers. We derived daughter colonies by serial dilution, verifying monoclonality using the Humara assay. hFLMPCs, which have been maintained in culture for up to 100 population doublings, have a high self-renewal capability with a doubling time of 46 h. The immunophenotype is: CD34+, CD90+, c-kit+, EPCAM+, c-met+, SSEA-4+, CK18+, CK19+, albumin-, alpha-fetoprotein-, CD44h+, and vimentin+. Passage 1 (P1) and P10 cells have identical morphology, immunophenotype, telomere length, and differentiation capacity. Placed in appropriate media, hFLMPCs differentiate into hepatocytes and bile duct cells, as well as into fat, bone, cartilage, and endothelial cells. Our results suggest that hFLMPCs are mesenchymal-epithelial transitional cells, probably derived from mesendoderm. hFLMPCs survive and differentiate into functional hepatocytes in vivo when transplanted into animal models of liver disease. hFLMPCs are a valuable tool for the study of human liver development, liver injury, and hepatic repopulation.
Collapse
Affiliation(s)
| | - K. J. Riehle
- Departments of *Pathology and
- Surgery, University of Washington, Seattle, WA 98115
| | | | - N. Teoh
- Departments of *Pathology and
| | | | | | - N. Fausto
- Departments of *Pathology and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
66
|
Baum C, Schambach A, Bohne J, Galla M. Retrovirus Vectors: Toward the Plentivirus? Mol Ther 2006; 13:1050-63. [PMID: 16632409 DOI: 10.1016/j.ymthe.2006.03.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/20/2005] [Revised: 03/16/2006] [Accepted: 03/16/2006] [Indexed: 01/19/2023] Open
Abstract
Recombinant retroviral vectors based upon simple gammaretroviruses, complex lentiviruses, or potentially nonpathogenic spumaviruses represent relatively well characterized tools that are widely used for stable gene transfer. Different members of the Retroviridae family have developed distinct and potentially useful features related to their life cycle. These natural differences can be exploited for specialized applications in gene therapy and could conceivably be combined to create future retroviral hybrid vectors, ideally incorporating the following features: an efficient, noncytopathic packaging system with low likelihood of recombination; serum resistance; an ability to pseudotype with cell-specific envelopes; high-fidelity reverse transcription before cell entry; unrestricted cytoplasmic transport and nuclear import; an insulated expression cassette; specific chromosomal targeting; and physiologic or regulated levels of transgene expression. We envisage that, compared to contemporary vectors, a hybrid vector combining these properties would have increased therapeutic efficacy and an enhanced biosafety profile. Many of the above goals will require the inclusion of nonretroviral components into vector particles or transgenes.
Collapse
Affiliation(s)
- Christopher Baum
- Department of Experimental Hematology, Hannover Medical School, D-30625 Hannover, Germany.
| | | | | | | |
Collapse
|
67
|
Dullaers M, Thielemans K. From pathogen to medicine: HIV-1-derived lentiviral vectors as vehicles for dendritic cell based cancer immunotherapy. J Gene Med 2006; 8:3-17. [PMID: 16288497 DOI: 10.1002/jgm.846] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/15/2022] Open
Abstract
Over the years, the unique capacity of dendritic cells (DC) for efficient activation of naive T cells has led to their extensive use in cancer immunotherapy protocols. In order to be able to fulfil their role as antigen-presenting cells, the antigen of interest needs to be efficiently introduced and subsequently correctly processed and presented by the DC. For this purpose, a variety of both viral and non-viral antigen-delivery systems have been evaluated. Amongst those, HIV-1-derived lentiviral vectors have been used successfully to transduce DC. This review considers the use of HIV-1-derived lentiviral vectors to transduce human and murine DC for cancer immunotherapy. Lentivirally transduced DC have been shown to present antigenic peptides, prime transgene-specific T cells in vitro and elicit a protective cytotoxic T-lymphocyte (CTL) response in animal models. Different parameters determining the efficacy of transduction are considered. The influence of lentiviral transduction on the DC phenotype and function is described and the induction of immune responses by lentivirally transduced DC in vitro and in vivo is discussed in detail. In addition, direct in vivo administration of lentiviral vectors aiming at the induction of antigen-specific immunity is reviewed. This strategy might overcome the need for ex vivo generation and antigen loading of DC. Finally, future perspectives towards the use of lentiviral vectors in cancer immunotherapy are presented.
Collapse
Affiliation(s)
- Melissa Dullaers
- Laboratory of Molecular and Cellular Therapy, Department of Physiology-Immunology, Medical School of the Vrije Universiteit Brussel (VUB), Laarbeeklaan 103/E, 1090 Brussels, Belgium
| | | |
Collapse
|
68
|
Goddard G, Martin JC, Naivar M, Goodwin PM, Graves SW, Habbersett R, Nolan JP, Jett JH. Single particle high resolution spectral analysis flow cytometry. Cytometry A 2006; 69:842-51. [PMID: 16969803 DOI: 10.1002/cyto.a.20320] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND While conventional multiparameter flow cytometers have proven highly successful, there are several types of analytical measurements that would benefit from a more comprehensive and flexible approach to spectral analysis including, but certainly not limited to spectral deconvolution of overlapping emission spectra, fluorescence resonance energy transfer measurements, metachromic dye analysis, free versus bound dye resolution, and Raman spectroscopy. METHODS Our system utilizes a diffraction grating to disperse the collected fluorescence and side-scattered light from cells or microspheres passing through the interrogation region over a rectangular charge-coupled-device image sensor. The flow cell and collection optics are taken from a conventional flow cytometer with minimal modifications to assure modularity of the system. RESULTS Calibration of the prototype spectral analysis flow cytometer included wavelength characterization and calibration of the dispersive optics. Benchmarking of the system demonstrated a single particle/cell intensity sensitivity of 2160 MESF of R-Phycoerythrin. Single particle spectra taken with our instrument were validated against bulk solution fluorimeter and conventional flow cytometer measurements. Coefficients of variation of integrated spectral fluorescence intensity of several sets of standard fluorescent microspheres ranged from 1.4 to 4.8% on the spectral system. Spectral discrimination of free versus PI bound to cells is also demonstrated. CONCLUSIONS It is demonstrated that the flow spectrometer has sufficient sensitivity and wavelength resolution to detect single cells and microspheres, including multi-fluorophore labeled microspheres. The capability to use both standard mathematical deconvolution techniques for data analysis, coupled with the feasibility of integration with existing flow cytometers, will improve the accuracy and precision of ratiometric measurements, enable the analysis of more discrete emission bands within a given wavelength range, and allow more precise resolution of the relative contribution of individual fluorophores in multiply-tagged samples, thereby enabling a range of new applications involving the spectral analysis of single cells and particles.
Collapse
Affiliation(s)
- Gregory Goddard
- National Flow Cytometry Resource, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 87545, USA.
| | | | | | | | | | | | | | | |
Collapse
|
69
|
Abstract
The host range of retroviral vectors including lentiviral vectors can be expanded or altered by a process known as pseudotyping. Pseudotyped lentiviral vectors consist of vector particles bearing glycoproteins (GPs) derived from other enveloped viruses. Such particles possess the tropism of the virus from which the GP was derived. For example, to exploit the natural neural tropism of rabies virus, vectors designed to target the central nervous system have been pseudotyped using rabies virus-derived GPs. Among the first and still most widely used GPs for pseudotyping lentiviral vectors is the vesicular stomatitis virus GP (VSV-G), due to the very broad tropism and stability of the resulting pseudotypes. Pseudotypes involving VSV-G have become effectively the standard for evaluating the efficiency of other pseudotypes. This review samples a few of the more prominent examples from the ever-expanding list of published lentiviral pseudotypes, noting comparisons made with pseudotypes involving VSV-G in terms of titer, viral particle stability, toxicity, and host-cell specificity. Particular attention is paid to publications of successfully targeting a specific organ or cell types.
Collapse
Affiliation(s)
- James Cronin
- Gene Therapy Program, Louisiana State University Health Sciences Center, New Orleans, 70112, USA
| | | | | |
Collapse
|
70
|
Du X, Qiu B, Zhan X, Kolmakova A, Gao F, Hofmann LV, Cheng L, Chatterjee S, Yang X. Radiofrequency-enhanced vascular gene transduction and expression for intravascular MR imaging-guided therapy: feasibility study in pigs. Radiology 2005; 236:939-44. [PMID: 16040894 DOI: 10.1148/radiol.2363041021] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/11/2022]
Abstract
PURPOSE To evaluate the feasibility of radiofrequency (RF)-enhanced vascular gene transduction and expression by using a magnetic resonance (MR) imaging-heating guidewire as an intravascular heating vehicle during MR imaging-guided therapy. MATERIALS AND METHODS The institutional committee for animal care and use approved the experimental protocol. The study included in vitro evaluation of the use of RF energy to enhance gene transduction and expression in vascular cells, as well as in vivo validation of the feasibility of intravascular MR imaging-guided RF-enhanced vascular gene transduction and expression in pig arteries. For in vitro experiments, approximately 10(4) vascular smooth muscle cells were seeded in each of four chambers of a cell culture plate. Next, 1 mL of a green fluorescent protein gene (gfp)-bearing lentivirus was added to each chamber. Chamber 4 was heated at approximately 41 degrees C for 15 minutes by using an MR imaging-heating guidewire connected to a custom RF generator. At day 6 after transduction, the four chambers were examined and compared at confocal microscopy to determine the efficiency of gfp transduction and expression. For the in vivo experiments, a lentivirus vector bearing a therapeutic gene, vascular endothelial growth factor 165 (VEGF-165), was transferred by using a gene delivery balloon catheter in 18 femoral-iliac arteries (nine artery pairs) in domestic pigs and Yucatan pigs with atherosclerosis. During gene infusion, one femoral-iliac artery in each pig was heated to approximately 41 degrees C with RF energy transferred via the intravascular MR imaging-heating guidewire, while the contralateral artery was not heated (control condition). At day 6, the 18 arteries were harvested for quantitative Western blot analysis to compare VEGF-165 transduction and expression efficiency between RF-heated and nonheated arterial groups. RESULTS Confocal microscopy showed gfp expression in chamber 4 that was 293% the level of expression in chamber 1 (49.6% +/- 25.8 vs 16.8% +/- 8.0). Results of Western blot analysis showed VEGF-165 expression for normal arteries in the RF-heated group that was 300% the level of expression in the nonheated group (70.4 arbitrary units [au] +/- 107.1 vs 23.5 au +/- 29.8), and, for atherosclerotic arteries in the RF-heated group, 986% the level in the nonheated group (129.2 au +/- 100.3 vs 13.1 au +/- 4.9). CONCLUSION Simultaneous monitoring and enhancement of vascular gene delivery and expression is feasible with the MR imaging-heating guidewire.
Collapse
Affiliation(s)
- Xiangying Du
- Department of Radiology, Johns Hopkins University School of Medicine, Traylor Bldg, Room 330, 720 Rutland Ave, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Dravid G, Ye Z, Hammond H, Chen G, Pyle A, Donovan P, Yu X, Cheng L. Defining the role of Wnt/beta-catenin signaling in the survival, proliferation, and self-renewal of human embryonic stem cells. Stem Cells 2005; 23:1489-501. [PMID: 16002782 DOI: 10.1634/stemcells.2005-0034] [Citation(s) in RCA: 257] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/31/2022]
Abstract
We used a panel of human and mouse fibroblasts with various abilities for supporting the prolonged growth of human embryonic stem cells (hESCs) to elucidate growth factors required for hESC survival, proliferation, and maintenance of the undifferentiated and pluripotent state (self-renewal). We found that supportive feeder cells secrete growth factors required for both hESC survival/proliferation and blocking hESC spontaneous differentiation to achieve self-renewal. The antidifferentiation soluble factor is neither leukemia inhibitory factor nor Wnt, based on blocking experiments using their antagonists. Because Wnt/beta-catenin signaling has been implicated in cell-fate determination and stem cell expansion, we further examined the effects of blocking or adding recombinant Wnt proteins on undifferentiated hESCs. In the absence of feeder cell-derived factors, hESCs cultured under a feeder-free condition survived/proliferated poorly and gradually differentiated. Adding recombinant Wnt3a stimulated hESC proliferation but also differentiation. After 4-5 days of Wnt3a treatment, hESCs that survived maintained the undifferentiated phenotype but few could form undifferentiated hESC colonies subsequently. Using a functional reporter assay, we found that the beta-catenin-mediated transcriptional activation in the canonical Wnt pathway was minimal in undifferentiated hESCs, but greatly upregulated during differentiation induced by the Wnt treatment and several other methods. Thus, Wnt/beta-catenin activation does not suffice to maintain the undifferentiated and pluripotent state of hESCs. We propose a new model for the role of Wnt/beta-catenin signaling in undifferentiated hESCs.
Collapse
Affiliation(s)
- Gautam Dravid
- The Institute for Cell Engineering, Department of Gynecology & Obstetrics, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | |
Collapse
|
72
|
Chen K, Chen L, Zhao P, Marrero L, Keoshkerian E, Ramsay A, Cui Y. FL-CTL assay: fluorolysometric determination of cell-mediated cytotoxicity using green fluorescent protein and red fluorescent protein expressing target cells. J Immunol Methods 2005; 300:100-14. [PMID: 15899496 DOI: 10.1016/j.jim.2005.02.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/09/2004] [Revised: 02/24/2005] [Accepted: 02/28/2005] [Indexed: 10/25/2022]
Abstract
Cytotoxic T lymphocytes (CTLs) are crucial effectors against intracellular pathogens and cancer. Accurate and efficient assessment of CTL activity is important for basic and clinical studies. Widely used CTL assays, including the chromium release, JAM test and ELISPOT, involve either radioisotopes or lengthy procedures. Here, we developed a new fluorolysometric CTL assay based on cell-mediated cytolysis of fluorescent protein (GFP or DsRed) expressing cells quantified by one of the fluoro-based methods: flow cytometry, fluorescence microplate reader, or fluorescence microscopy. With flexible detection methods and lentiviral vector transduced stable lines of either GFP+ or DsRed+ cells as targets for antigen presentation and equal number of the other as internal reference for consistency and accuracy, this assay is easy to perform and to scale-up for simultaneous multi-sample analyses. Using two different antigen systems, we demonstrated that this assay is very sensitive to determine primary CTL activity of both in vitro and in vivo primed antigen-specific T cells. Thus, this FL-CTL assay is highly sensitive, reliable, reproducible, economical, convenience and supports broad applications compared to conventional CTL assays.
Collapse
Affiliation(s)
- Kong Chen
- Louisiana State University Health Sciences Center, Gene Therapy Program, Department of Genetics and Medicine, 533 Bolivar Street, New Orleans, LA 70112, USA
| | | | | | | | | | | | | |
Collapse
|
73
|
Martín F, Toscano MG, Blundell M, Frecha C, Srivastava GK, Santamaría M, Thrasher AJ, Molina IJ. Lentiviral vectors transcriptionally targeted to hematopoietic cells by WASP gene proximal promoter sequences. Gene Ther 2005; 12:715-23. [PMID: 15750617 DOI: 10.1038/sj.gt.3302457] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/31/2023]
Abstract
The development of vectors that express a therapeutic transgene efficiently and specifically in hematopoietic cells (HCs) is an important goal for gene therapy of hematological disorders. In order to achieve this, we used a 500 bp fragment from the proximal WASP gene promoter to drive the expression of the WASP cDNA in the context of a self-inactivating lentiviral vector. Single-round transduction of WASp-deficient herpesvirus saimiri (HVS)-immortalized cells as well as primary allospecific T cells from Wiskott-Aldrich syndrome (WAS) patients with this vector (WW) resulted in expression levels similar to those of control cells. Non-HCs were transduced with similar efficiency, but the levels of WASp were 135-350 times lower than those achieved in HCs. Additionally, transduction of WASp-deficient cells with WW conferred a selective growth advantage in vitro. Therefore, lentiviral vectors incorporating proximal promoter sequences from the WASP gene confer hematopoietic-specific, and physiological protein expression.
Collapse
Affiliation(s)
- F Martín
- IPB 'López Neyra' CSIC, Granada, Spain
| | | | | | | | | | | | | | | |
Collapse
|
74
|
Cravens PD, Melkus MW, Padgett-Thomas A, Islas-Ohlmayer M, Del P Martin M, Garcia JV. Development and Activation of Human Dendritic Cells In Vivo in a Xenograft Model of Human Hematopoiesis. Stem Cells 2005; 23:264-78. [PMID: 15671149 DOI: 10.1634/stemcells.2004-0116] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/17/2022]
Abstract
Dendritic cells (DCs) are derived from CD34+ progenitors and play a central role in the development of immune responses and in tolerance. Their therapeutic potential underscores the need for in vivo models that accurately recapitulate human DC development and function to provide a better understanding of DC biology in health and disease. Using nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice transplanted with human CD34+ cells as a model of human hematopoiesis, we examined DC ontogeny. Progenitors of both myeloid (m) and plasmacytoid (p) DCs were identified in the bone marrow of mice up to 24 weeks after transplant, indicating ongoing and sustained production of DCs after initial engraftment. To determine whether human DCs derived from transplanted stem cells were functional, their response to acute inflammation using lipopolysaccharide (LPS) was examined. Eighteen hours after LPS administration, a dramatic increase in the plasma levels of the human inflammatory cytokines interleukin (IL)-8, IL-10, tumor necrosis factor-alpha, and IL-12p70 was observed. Only mDCs and not pDCs responded in vivo to LPS by upregulating CD86 and CD83. In vivo activation of human mDCs resulted in a substantial increase in the ability of mDCs to induce the proliferation of naive human T cells. Taken together, these data indicate that human CD34+ cells seem to have differentiated appropriately within the NOD/SCID microenvironment into DCs that are developmentally, phenotypically, and functionally similar to the DC subsets found in humans.
Collapse
Affiliation(s)
- Petra D Cravens
- Division of Infectious Diseases Y9.206, Department of Internal Medicine, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390-9113, USA
| | | | | | | | | | | |
Collapse
|
75
|
Moreau T, Bardin F, Imbert J, Chabannon C, Tonnelle C. Restriction of transgene expression to the B-lymphoid progeny of human lentivirally transduced CD34+ cells. Mol Ther 2005; 10:45-56. [PMID: 15233941 DOI: 10.1016/j.ymthe.2004.04.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/13/2004] [Accepted: 04/05/2004] [Indexed: 11/16/2022] Open
Abstract
Development of gene transfer strategies will necessitate improved efficiency and control of transduction and transgene expression. We here provide evidence that targeting expression of the GFP reporter gene to the B-lymphoid progeny of genetically modified human hematopoietic progenitor cells can be achieved through the insertion of regulatory sequences from the human CD19 gene promoter into a lentiviral vector. Based on a bioinformatics approach, three human CD19-derived sequences were designed and inserted into a self-inactivated lentiviral vector backbone upstream of the GFP gene: S.CD19 (230 bp), M.CD19 (464 bp), and L.CD19 (1274 bp). These new lentiviral vectors efficiently transduced cord blood CD34(+) cells. The M.CD19 and especially L.CD19 sequences preferentially targeted GFP expression to in vitro and in vivo differentiated CD19(+) progeny; moreover, transgene expression was detected from the CD34(+) pro/pre-B cell to the mature peripheral IgM(+) B cell stage. In contrast, GFP expression was weak or absent in primary T-lymphoid and uncommitted progenitor cells or in erythroid, natural killer, or myeloid differentiated cells. Such B-lineage-specific lentiviral vectors may be useful for correcting inherited disorders that affect B-lymphoid cells or for deciphering the transcriptional program that controls B cell commitment and differentiation.
Collapse
Affiliation(s)
- Thomas Moreau
- Centre de Thérapie Cellulaire et Génique, Institut Paoli-Calmettes, Centre Régional de Lutte contre le Cancer Provence-Alpes-Côte d'Azur, Marseille, France
| | | | | | | | | |
Collapse
|
76
|
Dijon M, Torne-Celer C, Moreau T, Tonnelle C, Chabannon C. Expression and recombination of the EGFP and EYFP genes in lentiviral vectors carrying two heterologous promoters. Cytotherapy 2005; 7:417-26. [PMID: 16236631 DOI: 10.1080/14653240500319317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Expressing two genes in the progeny of stem and progenitor cells that are transduced with a unique viral vector is desirable in certain situations. We tested the ability of two lentiviral vectors to transduce human cells of hematopoietic origin and concomitantly express two reporter genes, either EGFP (enhanced green fluorescent protein) and DsRed2, or EGFP and EYFP (enhanced yellow fluorescent protein), from two internal promoters. METHODS The vectors were generated from the pTRIP deltaU3 EF1alpha EGFP lentiviral vector. Following transduction of hematopoietic and non-hematopoietic cell lines, we performed FACS, PCR and Southern blot analyzes to quantify transduction, integration efficiencies and size of integrated lentiviral vectors, respectively. RESULTS The detection of DsRed2 fluorescence appeared unexpectedly low in human cells of hematopoietic origin. Alternatively, a modification in the flow cytometry assay allowed us to distinguish between the two overlapping fluorescence signals emitted by EGFP and EYFP, when transduced cells were excited with a 488-nm laser beam. However, the low frequency of double-positive EGFP+ EYFP+ cells, and the existence of single-positive, mostly EGFP- EYFP+, cells, prompted us to search for recombinations in the vector sequence. Southern blotting of DNA obtained from transduced cells indeed demonstrated that recombination had occurred between the two closely related EGFP and EYFP sequences. DISCUSSION These observations suggest that recombination occurred within the EGFP and EYFP genes, which differ by only four amino acids. We conclude that the insertion of two highly homologous sequences into a lentiviral backbone can favor recombination.
Collapse
Affiliation(s)
- M Dijon
- Centre de Thérapie Cellulaire et Génique, Institut Paoli-Calmettes, Centre Régional de Lutte Contre le Cancer Provence-Alpes-Côte d'Azur, and Unité Mixte de Recherche 599, Institut de Recherche sur le Cancer de Marseille, Marseille, France
| | | | | | | | | |
Collapse
|
77
|
Lee JM, Lee KH, Farrell CJ, Ling PD, Kempkes B, Park JH, Hayward SD. EBNA2 is required for protection of latently Epstein-Barr virus-infected B cells against specific apoptotic stimuli. J Virol 2004; 78:12694-7. [PMID: 15507659 PMCID: PMC525073 DOI: 10.1128/jvi.78.22.12694-12697.2004] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/22/2022] Open
Abstract
In addition to functioning as a transcriptional transactivator, Epstein-Barr virus EBNA2 interacts with Nur77 to protect against Nur77-mediated apoptosis. Estrogen-regulated EBNA2 in EREB2-5 cells was replaced by either EBNA2 or EBNA2 with a deletion of conserved region 4 (EBNA2DeltaCR4). Both EBNA2-converted and EBNA2DeltaCR4-converted EREB2-5 cells grew in the absence of estrogen and expressed LMP1. Treatment with tumor necrosis factor alpha did not induce apoptosis of EBNA2- or EBNA2DeltaCR4-expressing cells, but EBNA2DeltaCR4 cells were susceptible to etoposide and 5-fluorouracil, Nur77-mediated inducers of apoptosis. Thus, EBNA2 protects B cells against specific apoptotic agents against which LMP1 is not effective.
Collapse
Affiliation(s)
- Jae Myun Lee
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, School of Medicine, Bunting-Blaustein Building CRB308, 1650 Orleans Street, Baltimore, MD 21231, USA
| | | | | | | | | | | | | |
Collapse
|
78
|
Chen HH, Zhan X, Kumar A, Du X, Hammond H, Cheng L, Yang X. Detection of dual-gene expression in arteries using an optical imaging method. JOURNAL OF BIOMEDICAL OPTICS 2004; 9:1223-9. [PMID: 15568943 DOI: 10.1117/1.1803842] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 05/24/2023]
Abstract
We evaluate the in vivo use of an optical imaging method to detect the vascular expression of green fluorescent protein (GFP) or red fluorescent protein (RFP), and to detect the simultaneous expression of GFP and RFP after transduction into arteries by a dual-promoter lentiviral vector driving their concurrent expression. This method involves using a charge-coupled device camera to detect fluorescence, a fiber optic probe to transmit light, and optical filters to distinguish each marker. In animal models, these vectors are locally delivered to target arteries, whereas the gene for a nonfluorescent cell-surface protein is transduced into contralateral arteries as the sham control. The images show distinct areas of bright fluorescence from GFP and RFP along the target arteries on excitation; no exogenous fluorescence is observed in the controls. Measured signal intensities from arteries transduced with the single- and dual-promoter vectors exceed the autofluorescence signal from the controls. Transgene expression of GFP and RFP in vivo is confirmed with confocal microscopy. We demonstrate the use of an optical imaging method to concurrently detect two distinct fluorescent proteins, potentially permitting the expression of multiple transgenes and their localization in the vasculature to be monitored.
Collapse
Affiliation(s)
- Hunter H Chen
- Johns Hopkins University, Department of Biomedical Engineering, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | |
Collapse
|
79
|
Abstract
Over the past two decades, the ability to transfer genes into hematopoietic stem cells (HSCs) has provided new insights into the behavior of individual stem cells and offered a novel approach for the treatment of various inherited or acquired disorders. At present, gene transfer into HSCs has been achieved mainly using modified retroviruses. While retrovirus-based vectors could efficiently transduce murine HSCs, extrapolation of these methods to large mammals and human clinical trials resulted in very low numbers of gene-marked engrafted cells. In addition, in vitro progenitor assays used to optimize gene transfer procedures were found to poorly predict the outcome of stem cell gene transfer. The focus rapidly turned to the development of superior and more relevant preclinical assays in human stem cell gene transfer research. Xenogeneic transplant models and large animal transplantation system have been invaluable. The development of better assays for evaluating human gene therapy protocols and a better understanding of stem cell and vector biology has culminated over the past decade in multiple strategies to improve gene transfer efficiency into HSCs. Improved gene transfer vectors, optimization of cytokine combination, and incorporation of a recombinant fragment of fibronectin during transduction are examples of novel successful additions to the early gene transfer protocols that have contributed to the first unequivocal clinical benefits resulting from genetic manipulation of HSC.
Collapse
Affiliation(s)
- André Larochelle
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
80
|
Li Z, Modlich U, Baum C. Safety and efficacy in retrovirally modified haematopoietic cell therapy. Best Pract Res Clin Haematol 2004; 17:493-503. [PMID: 15498719 DOI: 10.1016/j.beha.2004.05.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/17/2023]
Abstract
The enormous therapeutic potential of haematopoietic stem cells may be realized if we acquire the ability to control their survival in vitro and their behaviour in vivo. While extrinsic approaches using drugs are unlikely to be developed for this purpose in the near future, altering the intrinsic pool of biological information of stem cells by somatic gene transfer is more likely to succeed. Using the first generations of retroviral gene transfer vectors, we are confronted with the first examples of both successful therapeutic interventions and severe adverse events. The latter are related to the incomplete precision of the existing technologies. Concerted safety and efficiency evaluation has been enforced to further improve the prospects of this field. This review summarizes the current state of the debate, proposing future research directions towards understanding the complex interplay of risk factors related to random transgene insertion, unphysiological transgene expression and additional contributory factors of the specific therapeutic setting.
Collapse
Affiliation(s)
- Zhixiong Li
- Department of Haematology, Haemostaseology and Oncology, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany
| | | | | |
Collapse
|
81
|
Werner M, Kraunus J, Baum C, Brocker T. B-cell-specific transgene expression using a self-inactivating retroviral vector with human CD19 promoter and viral post-transcriptional regulatory element. Gene Ther 2004; 11:992-1000. [PMID: 15029232 DOI: 10.1038/sj.gt.3302255] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/23/2023]
Abstract
Retroviral gene transfer resulting in transgene expression selectively restricted to specific cell lineages would be desirable for many gene therapeutic applications. Such transcriptional targeting of retroviruses can be accomplished by employing eukaryotic control elements in self-inactivating (SIN) retroviral vectors, but use of these vectors is complicated by an accompanying reduction in viral titers. To overcome this restriction and address the influence of the post-transcriptional regulatory element of the Woodchuck hepatitis virus (WPRE) on viral titers and transgene expression, we developed SIN-vectors with and without WPRE. Using the enhancer-promoter of the Spleen Focus Forming virus (SFFV) to direct eGFP expression to multiple hematopoietic lineages, we show that WPRE significantly (>10 x) increased viral titers (>10(6) per ml of unconcentrated supernatant) and transgene expression in NIH3T3 cells in vitro. Gene expression in vivo was significantly lowered in lymphoid cells, but not in myeloid cells when WPRE was present. Furthermore, the use of WPRE in combination with the B-cell lineage-specific CD19 promoter significantly increased viral titers and allowed targeting of transgene expression by SIN-vectors specifically to B cells throughout their development in primary and secondary lymphoid organs.
Collapse
Affiliation(s)
- M Werner
- Institute for Immunology, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | |
Collapse
|
82
|
Zhan X, Dravid G, Ye Z, Hammond H, Shamblott M, Gearhart J, Cheng L. Functional antigen-presenting leucocytes derived from human embryonic stem cells in vitro. Lancet 2004; 364:163-71. [PMID: 15246729 DOI: 10.1016/s0140-6736(04)16629-4] [Citation(s) in RCA: 122] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Differentiated cells derived from pluripotent human embryonic stem (hES) cells offer the opportunity for new transplantation therapies. However, hES cells and their differentiated progeny express highly polymorphic MHC molecules that serve as major graft rejection antigens to the immune system of allogeneic hosts. To achieve sustained engraftment of donor cells, strategies must be developed to overcome graft rejection without broadly suppressing host immunity. One approach entails induction of donor-specific immune tolerance by establishing chimeric engraftment in hosts with haemopoietic cells derived from an existing hES cell line. We aimed to develop methods to efficiently differentiate hES cells to haemopoietic cells, including immune-modulating leucocytes, a prerequisite of the tolerance induction strategies applying to hES cell-mediated transplantation. METHODS We developed a method to generate a broad range of haemopoietic cells from hES-generated embryonic bodies in the absence of murine stromal feeder cells. Embryonic bodies were further cultured in the presence of haemopoietic cytokines. In addition to flow cytometric analyses of haemopoietic cell markers, we analysed the hES cell-derived haemopoietic cells by colony-forming assays (for erythroid and myeloid progenitor cells), cytochemical staining, and mixed leucocyte reactions to determine the functional capacity of the generated antigen-presenting cells. FINDINGS 12 independent experiments were done. When selected growth factors were added, leucocytes expressing CD45 were generated and released into culture media for 6-7 weeks. Under the condition used, both erythroid and myeloid progenitor cells were generated. About 25% of the generated leucocytes acquired MHC class II and costimulatory molecule expression. These hES-derived, MHC class II+ leucocytes resembled dendritic cells and macrophages, and they functioned as antigen-presenting cells capable of eliciting allogeneic CD4 and CD8 T-cell responses in culture. INTERPRETATION The hES cell-derived antigen-presenting cells could be used to regulate alloreactive T cells and induce immune tolerance for improvement of the transplant acceptance of hES-cell derivatives.
Collapse
Affiliation(s)
- Xiangcan Zhan
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | |
Collapse
|
83
|
Abstract
Despite the remarkable progress achieved in the treatment of leukemias over the last several years, many problems (multidrug resistance [MDR], cellular heterogeneity, heterogeneous molecular abnormalities, karyotypic instability, and lack of selective action of antineoplastic agents) still remain. The recent progress in tumor molecular biology has revealed that leukemias are likely to arise from disruption of differentiation of early hematopoietic progenitors that fail to give birth to cell lineage restricted phenotypes. Evidence supporting such mechanisms has been derived from studying bone marrow leukemiogenesis and analyzing differentiation of leukemic cell lines in culture that serve as models of erythroleukemic (murine erythroleukemia [MEL] and human leukemia [K562] cells) and myeloid (human promyelocytic leukemia [HL-60] cells) cell maturation. This paper reviews the current concepts of differentiation, the chemical/pharmacological inducing agents developed thus far, and the mechanisms involved in initiation of leukemic cell differentiation. Emphasis was given on commitment and the cell lineage transcriptional factors as key regulators of terminal differentiation as well as on membrane-mediated events and signaling pathways involved in hematopoietic cell differentiation. The developmental program of MEL cells was presented in considerable depth. It is quite remarkable that the erythrocytic maturation of these cells is orchestrated into specific subprograms and gene expression patterns, suggesting that leukemic cell differentiation represents a highly coordinated set of events that lead to irreversible growth arrest and expression of cell lineage restricted phenotypes. In MEL and other leukemic cells, differentiation appears to be accompanied by differentiation-dependent apoptosis (DDA), an event that can be exploited chemotherapeutically. The mechanisms by which the chemical inducers promote differentiation of leukemic cells have been discussed.
Collapse
Affiliation(s)
- Asterios S Tsiftsoglou
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, Aristotle University of Thessaloniki, Thessaloniki, GR-54124, Greece.
| | | | | |
Collapse
|
84
|
Dunlap S, Yu X, Cheng L, Civin CI, Alani RM. High-efficiency stable gene transduction in primary human melanocytes using a lentiviral expression system. J Invest Dermatol 2004; 122:549-51. [PMID: 15009744 DOI: 10.1046/j.0022-202x.2004.22214.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/20/2022]
|
85
|
|
86
|
Pan F, Ye Z, Cheng L, Liu JO. Myocyte enhancer factor 2 mediates calcium-dependent transcription of the interleukin-2 gene in T lymphocytes: a calcium signaling module that is distinct from but collaborates with the nuclear factor of activated T cells (NFAT). J Biol Chem 2004; 279:14477-80. [PMID: 14722108 DOI: 10.1074/jbc.c300487200] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/06/2022] Open
Abstract
The second messenger calcium plays an essential role in the T cell receptor-mediated signal transduction pathways leading to transcription of the interleukin-2 gene. A key mechanism of calcium signaling has been shown to be mediated by calcineurin and NFAT. We report herein that the transcription factor myocyte enhancer factor (MEF)-2 is another calcium signal transducer involved in the regulation of the interleukin (IL)-2 promoter. A MEF2-binding site was identified in proximity to the TATA box of the IL-2 promoter. This site was shown to be bound by MEF2 in both resting and activated T cells. Overexpression of MEF2 enhanced, while overexpression of a dominant negative form of MEF2 or the MEF2-specific transcriptional corepressors Cabin1 and histone deacetylase 4 inhibited, the T cell receptor-dependent activation of an IL-2 reporter gene. Down-regulation of MEF2 by RNA interference in primary human T cells led to the inhibition of endogenous IL-2 transcription. These results suggest that MEF2 is required for the transcriptional activation of IL-2 and likely other cytokine genes in response to calcium signaling and may serve as a novel target for development of immunosuppressants.
Collapse
Affiliation(s)
- Fan Pan
- Department of Pharmacology and Molecular Science and Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | |
Collapse
|
87
|
Breckpot K, Heirman C, Neyns B, Thielemans K. Exploiting dendritic cells for cancer immunotherapy: genetic modification of dendritic cells. J Gene Med 2004; 6:1175-88. [PMID: 15468193 DOI: 10.1002/jgm.615] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/31/2022] Open
Abstract
Dendritic cells (DCs) are pivotal regulators of immune reactivity and immune tolerance. The observation that DCs can recruit naive T cells has invigorated cancer immunology and led to the proposal of DCs as the basis for vaccines designed for the treatment of cancer. Designing effective strategies to load DCs with antigens is a challenging field of research. The successful realization of gene transfer to DCs will be highly dependent on the employed vector system. Here, we review various viral and non-viral gene transfer systems, and discuss their distinct characteristics and possible advantages and disadvantages in respect to their use in DC-based immunotherapy.
Collapse
Affiliation(s)
- Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Department of Physiology and Immunology, Medical School of the Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | | | | | | |
Collapse
|
88
|
Abstract
PURPOSE OF REVIEW Atherosclerosis is a chronic inflammatory disease that is the primary cause of morbidity and mortality in the developed world. Many studies have shown that macrophages and T-cells play critical roles in multiple aspects of the pathogenesis of the disease. Given that these cells are ultimately derived from bone marrow precursors, the concept of performing gene therapy for atherosclerosis through the retroviral transduction of hematopoietic stem cells has received much attention. This review will highlight recent advances that will help bring this goal closer. RECENT FINDINGS The clinical application of retroviral gene transfer into hematopoietic stem cells has been hampered, in part, by the absence of vectors that can direct long-lasting, cell-type specific gene expression. In this review we will detail recent developments in the design of novel retroviral and lentiviral vectors that appear to overcome these problems, offering approaches to express therapeutic genes in specific cell-types within atherosclerotic lesions. We will also highlight advances in our understanding of the pathogenesis of atherosclerosis that may offer new gene therapeutic targets. SUMMARY The use of retroviral transduction of hematopoietic stem cells for treatment of patients with atherosclerosis still remains a long-term goal. However, the recent development of retroviral vectors capable of directing expression to specific cell types within the lesion will allow more targeted therapeutic strategies to be devised. In addition, these vectors will provide powerful experimental tools to further our understanding of the pathogenesis of the disease.
Collapse
Affiliation(s)
- Peter J Gough
- Department of Pathology, University of Washington, Harborview Medical Center, Seattle, Washington 98104-2499, USA.
| | | |
Collapse
|
89
|
Zhou X, Cui Y, Huang X, Yu Z, Thomas AM, Ye Z, Pardoll DM, Jaffee EM, Cheng L. Lentivirus-mediated gene transfer and expression in established human tumor antigen-specific cytotoxic T cells and primary unstimulated T cells. Hum Gene Ther 2003; 14:1089-105. [PMID: 12885348 DOI: 10.1089/104303403322124800] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/13/2022] Open
Abstract
In this report, we evaluated the efficiency of stable gene transfer into established CD8(+) human tumor antigen-specific cytotoxic T cell (CTL) lines and peripheral blood lymphocytes (PBL) by oncoretroviral and lentiviral vectors. In the oncoretroviral vector, the green fluorescent protein (GFP) reporter gene was regulated by the murine stem cell virus (MSCV) promoter. In three human immunodeficiency virus type 1 (HIV-1)-based lentiviral vectors, the GFP transgene was regulated by either a chimeric MSCV/HIV-1 promoter, or cellular promoters from human housekeeping genes PGK and EF1 alpha. We found that several lines of proliferating tumor-specific CTL were poorly (=2%) transduced by the oncoretroviral vector that transduced Jurkat T cell line efficiently (=80%). In contrast, three lentiviral vectors transduced 38-63% of these proliferating CTL. More interestingly, all lentiviral vectors packaged without the HIV-1 accessory proteins transduced human bulk PBL and purified CD4(+) and CD8(+) lymphocyte subsets without prior stimulation. Detailed analysis indicated that the lentiviral vectors containing the EF1 alpha or PGK ubiquitous promoter can transduce unstimulated PBL and achieve low-level transgene expression in the absence of any T-cell activation. However, T-cell activation subsequent to the transduction of unstimulated PBL is required for high-level transgene expression. Transduced PBL expressing transgene delivered by the lentiviral vectors still preserved resting and naïve cell phenotypes. Taken together, prior T cell stimulation and HIV-1 accessory proteins are dispensable for lentivirus-mediated gene transfer into resting naïve and memory T lymphocytes. These results will have significant implications for the study of T-cell biology and for the improvement of clinical gene therapies of acquired immune deficiency syndrome (AIDS) and cancer.
Collapse
Affiliation(s)
- Xianzheng Zhou
- Division of Immunology and Hematppoiesis, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Dingli D, Diaz RM, Bergert ER, O'Connor MK, Morris JC, Russell SJ. Genetically targeted radiotherapy for multiple myeloma. Blood 2003; 102:489-96. [PMID: 12649158 DOI: 10.1182/blood-2002-11-3390] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/03/2023] Open
Abstract
Multiple myeloma is a disseminated neoplasm of terminally differentiated plasma cells that is incurable with currently available therapies. Although the disease is radiosensitive, external beam radiation leads to significant toxicity due to sensitive end-organ damage. Thus, genetic approaches for therapy are required. We hypothesized that the incorporation of immunoglobulin promoter and enhancer elements in a self-inactivating (SIN) lentiviral vector should lead to specific and high-level transgene expression in myeloma cells. A SIN lentivector with enhanced green fluorescent protein (EGFP) expression under the control of a minimal immunoglobulin promoter as well as the Kappa light chain intronic and 3' enhancers transduced myeloma cell lines with high efficiency (30%-90%). EGFP was expressed at a high level in myeloma cells but silent in all nonmyeloma cell lines tested compared with the cytomegalovirus (CMV) promoter/enhancer. Transduction of myeloma cells with the targeted vector coding for the human sodiumiodide symporter (hNIS) led to hNIS expression by these cells allowing them to concentrate radioiodine up to 18-fold compared with controls. Tumor xenografts in severe combined immunodeficiency mice expressing hNIS could be imaged using iodine-123 (123I) and shown to retain iodide for up to 48 hours. These tumor xenografts were completely eradicated by a single dose of the therapeutic isotope iodine-131 (131I) without evidence of recurrence up to 5 months after therapy. We conclude that lentivectors can be transcriptionally targeted for myeloma cells and the use of hNIS as a therapeutic gene for myeloma in combination with 131I needs further exploration.
Collapse
MESH Headings
- Animals
- Cytomegalovirus/genetics
- Defective Viruses/genetics
- Drug Delivery Systems
- Enhancer Elements, Genetic
- Genes, Immunoglobulin
- Genes, Reporter
- Genes, Synthetic
- Genetic Vectors/genetics
- Genetic Vectors/therapeutic use
- Green Fluorescent Proteins
- Humans
- Immunoglobulin kappa-Chains/genetics
- Introns/genetics
- Iodine Radioisotopes/pharmacokinetics
- Iodine Radioisotopes/therapeutic use
- Lentivirus/genetics
- Luminescent Proteins/genetics
- Mice
- Mice, SCID
- Multiple Myeloma/metabolism
- Multiple Myeloma/pathology
- Multiple Myeloma/radiotherapy
- Myeloma Proteins/genetics
- Promoter Regions, Genetic
- Radiopharmaceuticals/pharmacokinetics
- Radiopharmaceuticals/therapeutic use
- Recombinant Fusion Proteins/physiology
- Symporters/genetics
- Symporters/physiology
- Transcription, Genetic
- Transduction, Genetic
- Tumor Cells, Cultured/metabolism
- Tumor Cells, Cultured/transplantation
- Xenograft Model Antitumor Assays
Collapse
|
91
|
Lutzko C, Senadheera D, Skelton D, Petersen D, Kohn DB. Lentivirus vectors incorporating the immunoglobulin heavy chain enhancer and matrix attachment regions provide position-independent expression in B lymphocytes. J Virol 2003; 77:7341-51. [PMID: 12805432 PMCID: PMC164820 DOI: 10.1128/jvi.77.13.7341-7351.2003] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/20/2022] Open
Abstract
In the present studies we developed lentivirus vectors with regulated, consistent transgene expression in B lymphocytes by incorporating the immunoglobulin heavy chain enhancer (E micro ) with and without associated matrix attachment regions (E micro MAR) into lentivirus vectors. Incorporation of these fragments upstream of phosphoglycerate kinase (PGK) or cytomegalovirus promoters resulted in a two- to threefold increase in enhanced green fluorescent protein (EGFP) mean fluorescence intensity (MFI) in B-lymphoid but not T-lymphoid, myeloid, fibroblast, or carcinoma cell lines. A 1-log increase in EGFP expression was observed in B-lymphoid cells (but not myeloid cells) differentiated from human CD34(+) progenitors in vitro transduced with E micro - and E micro MAR-containing lentivectors. Lastly, we evaluated the expression from the E micro MAR element in mice 2 to 24 weeks posttransplant with transduced hematopoietic stem cells. In mice receiving vectors with the E micro and E micro MAR elements upstream of the PGK promoter, there was a 2- to 10-fold increase in EGFP expression in B cells (but not other cell types). Evaluation of the coefficient of variation of expression among different cell types demonstrated that consistent, position-independent transgene expression was observed exclusively in B cells transduced with the E micro MAR-containing vector and not other cells types or vectors. Proviral genomes with the E micro MAR element had increased chromatin accessibility, which likely contributed to the position independence of expression in B lymphocytes. In summary, incorporation of the E micro MAR element in lentivirus vectors resulted in enhanced, position-independent expression in primary B lymphocytes. These vectors provide a useful tool for the study of B-lymphocyte biology and the development of gene therapy for disorders affecting B lymphocytes, such as immune deficiencies.
Collapse
Affiliation(s)
- Carolyn Lutzko
- Division of Research Immunology and Bone Marrow Transplantation at Childrens Hospital Los Angeles, California 90027, USA
| | | | | | | | | |
Collapse
|
92
|
Abstract
Gene therapy is an exciting frontier in medicine today. Many genes have been shown to be useful for treatment of various vascular diseases, including chronic cardiac and limb ischemia syndromes, vasculoproliferative disorder, hypercholesterolemia, atherosclerosis, thrombosis, and hypertension. Precise delivery of genes into target vessels, efficient transfer of genes into vascular cells of the target, and prompt assessment of gene expression over time are three challenging tasks for successful vascular gene therapy. Thus, in vivo imaging methods that can be used to monitor gene delivery and localize gene expression are needed. Modern imaging techniques provide an opportunity to monitor and direct vascular gene therapy. Radiologists play a key role not only in developing and mastering endovascular genetic interventions but also in assessing the success of vascular gene therapy and directing further refinement of vascular gene therapy technology. This article provides an overview of the current status of imaging of vascular gene therapy.
Collapse
Affiliation(s)
- Xiaoming Yang
- Department of Radiology, Johns Hopkins University School of Medicine, Traylor Bldg, Rm 330, 720 Rutland Ave, Baltimore, MD 21205, USA.
| |
Collapse
|
93
|
Cui Y, Kelleher E, Straley E, Fuchs E, Gorski K, Levitsky H, Borrello I, Civin CI, Schoenberger SP, Cheng L, Pardoll DM, Whartenby KA. Immunotherapy of established tumors using bone marrow transplantation with antigen gene--modified hematopoietic stem cells. Nat Med 2003; 9:952-8. [PMID: 12778137 DOI: 10.1038/nm882] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/23/2003] [Accepted: 04/23/2003] [Indexed: 12/22/2022]
Abstract
A major focus of cancer immunotherapy is to develop strategies to induce T-cell responses through presentation of tumor antigens by dendritic cells (DCs). Current vaccines are limited in their ability to efficiently transfer antigens to DCs in vivo. Ex vivo-generated DCs can be efficiently loaded with antigen but after reinjection, few DCs traffic to secondary lymphoid organs, the critical sites for antigen presentation. To enhance efficiency and durability of antigen presentation by DCs, we transduced hematopoietic stem-progenitor cells (HSCs) with a model tumor antigen and then transplanted the gene-modified cells into irradiated recipient mice, which resulted in efficient expression of the transgene in a large proportion of donor derived DCs in lymphoid organs. The combination of bone marrow transplantation (BMT) using transduced HSCs, systemic agents that generate and activate DCs, and mature T-cell infusion resulted in substantial expansion and activation of antigen-specific T cells. This tripartite strategy provided potent antigen-specific immunotherapy for an aggressive established tumor.
Collapse
MESH Headings
- Animals
- Antigens/metabolism
- Bone Marrow Transplantation/methods
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD40 Antigens/genetics
- CD40 Antigens/immunology
- CD40 Antigens/metabolism
- CD8-Positive T-Lymphocytes/physiology
- Cells, Cultured
- Dendritic Cells/physiology
- Hemagglutination/genetics
- Hematopoietic Stem Cell Transplantation/methods
- Hematopoietic Stem Cells/physiology
- Immunotherapy, Adoptive/methods
- Interferon-gamma/metabolism
- Lentivirus/genetics
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/therapy
- Mice
- Mice, Inbred BALB C
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/therapy
- Spleen/cytology
- Survival Rate
- T-Lymphocytes/immunology
- T-Lymphocytes/physiology
- Transduction, Genetic
- Transplantation, Autologous
Collapse
Affiliation(s)
- Yan Cui
- Immunology & Hematopoiesis Division, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland 21231, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Ramezani A, Hawley TS, Hawley RG. Performance- and safety-enhanced lentiviral vectors containing the human interferon-beta scaffold attachment region and the chicken beta-globin insulator. Blood 2003; 101:4717-24. [PMID: 12586614 DOI: 10.1182/blood-2002-09-2991] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/20/2022] Open
Abstract
Retroviral vectors are the most efficient means of stable gene delivery to hematopoietic stem cells (HSCs). However, transgene expression from retroviral vectors is frequently subject to the negative influence of chromosomal sequences flanking the site of integration. Toward the development of autonomous transgene expression cassettes, we inserted the human interferon-beta scaffold attachment region (IFN-SAR) and the chicken beta-globin 5' DNase I hypersensitive site 4 (5'HS4) insulator both separately and together into a series of self-inactivating (SIN) lentiviral vector backbones. Transduced cells of the human CD34+ hematopoietic progenitor line KG1a-pooled populations as well as individual clones harboring single integrants--were analyzed for reporter expression during culture periods of up to 4 months. Vectors carrying both the 5'HS4 insulator and the IFN-SAR consistently outperformed control vectors without inserts as well as vectors carrying either element alone. The performance of a set of vectors containing the murine stem cell virus long terminal repeat as an internal promoter was subsequently assessed during in vitro monocytic differentiation of transduced primary human CD34+ cord blood cells. Similar to what was observed in the KG1a hematopoietic progenitor cell model, optimal reporter expression in primary monocytes was obtained with the vector bearing both regulatory elements. These findings indicate that the 5'HS4/IFN-SAR combination is particularly effective at maintaining open chromatin domains permissive for high-level transgene expression at early and late stages of hematopoietic development, and thus could be of utility in HSC-directed retroviral vector-mediated gene transfer applications.
Collapse
Affiliation(s)
- Ali Ramezani
- Hematopoiesis Department, Flow Cytometry Facility, American Red Cross, Rockville, MD 20855, USA
| | | | | |
Collapse
|
95
|
Yu X, Zhan X, D'Costa J, Tanavde VM, Ye Z, Peng T, Malehorn MT, Yang X, Civin CI, Cheng L. Lentiviral vectors with two independent internal promoters transfer high-level expression of multiple transgenes to human hematopoietic stem-progenitor cells. Mol Ther 2003; 7:827-38. [PMID: 12788657 DOI: 10.1016/s1525-0016(03)00104-7] [Citation(s) in RCA: 123] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/27/2022] Open
Abstract
Lentiviral vectors (LVs) offer several advantages over traditional oncoretroviral vectors. LVs efficiently transduce slowly dividing cells, including hematopoietic stem-progenitor cells (HSCs), resulting in stable gene transfer and expression. Additionally, recently developed self-inactivating (SIN) LVs allow promoter-specific transgene expression. For many gene transfer applications, transduction of more than one gene is needed. We obtained inconsistent results in our attempts to coexpress two transgenes linked by an internal ribosomal entry site (IRES) element in a single bicistronic LV transcript. In more than six bicistronic LVs we constructed containing a gene of interest followed by an IRES and the GFP reporter gene, GFP fluorescence was undetectable in transduced cells. We therefore investigated how to achieve consistent and efficient coexpression of two transgenes by LVs. In a SIN LV containing the elongation factor 1alpha promoter, we included a second promoter from cytomegalovirus, the phosphoglycerate kinase gene, or the HLA-DRalpha gene. Using a single LV containing two constitutive promoters, we achieved strong and sustained expression of both transgenes in transduced engrafting CD34(+) HSCs and their progeny, as well as in other human cell types. Thus, such dual-promoter LVs can coexpress multiple transgenes efficiently in a single target cell and will enable many gene transfer applications.
Collapse
Affiliation(s)
- Xiaobing Yu
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Department of Oncology, Baltimore, Maryland 21231, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Abstract
Gene therapy has been applied in a variety of experimental models of autoimmunity with some success. In this article, we outline recent developments in gene therapy vectors, discuss advantages and disadvantages of each, and highlight their recent applications in autoimmune models. We also consider progress in vector targeting and components for regulating transgene expression, which will both improve gene therapy safety and empower gene therapy to fullfil its potential as a therapeutic modality. In conclusion, we consider candidate vectors that satisfy requirements for application in the principal therapeutic strategies in which gene therapy will be applied to autoimmune conditions.
Collapse
Affiliation(s)
- D J Gould
- 1Bone & Joint Research Unit, Barts & The London, Queen Mary's Medical School, University of London, London, UK
| | | |
Collapse
|
97
|
Steptoe RJ, Ritchie JM, Harrison LC. Transfer of hematopoietic stem cells encoding autoantigen prevents autoimmune diabetes. J Clin Invest 2003; 111:1357-63. [PMID: 12727927 PMCID: PMC154439 DOI: 10.1172/jci15995] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/24/2002] [Accepted: 02/26/2003] [Indexed: 11/17/2022] Open
Abstract
Bone marrow or hematopoietic stem cell transplantation is a potential treatment for autoimmune disease. The clinical application of this approach is, however, limited by the risks associated with allogeneic transplantation. In contrast, syngeneic transplantation would be safe and have wide clinical application. Because T cell tolerance can be induced by presenting antigen on resting antigen-presenting cells (APCs), we reasoned that hematopoietic stem cells engineered to express autoantigen in resting APCs could be used to prevent autoimmune disease. Proinsulin is a major autoantigen associated with pancreatic beta cell destruction in humans with type 1 diabetes (T1D) and in autoimmune NOD mice. Here, we demonstrate that syngeneic transplantation of hematopoietic stem cells encoding proinsulin transgenically targeted to APCs totally prevents the development of spontaneous autoimmune diabetes in NOD mice. This antigen-specific immunotherapeutic strategy could be applied to prevent T1D and other autoimmune diseases in humans.
Collapse
Affiliation(s)
- Raymond J Steptoe
- Autoimmunity and Transplantation Division, The Walter and Eliza Hall Institute of Medical Research, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | | | | |
Collapse
|
98
|
Marodon G, Mouly E, Blair EJ, Frisen C, Lemoine FM, Klatzmann D. Specific transgene expression in human and mouse CD4+ cells using lentiviral vectors with regulatory sequences from the CD4 gene. Blood 2003; 101:3416-23. [PMID: 12511423 DOI: 10.1182/blood-2002-02-0578] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/20/2022] Open
Abstract
Achieving cell-specific expression of a therapeutic transgene by gene transfer vectors represents a major goal for gene therapy. To achieve specific expression of a transgene in CD4(+) cells, we have generated lentiviral vectors expressing the enhanced green fluorescent protein (eGFP) reporter gene under the control of regulatory sequences derived from the CD4 gene--a minimal promoter and the proximal enhancer, with or without the silencer. Both lentiviral vectors could be produced at high titers (more than 10(7) infectious particles per milliliter) and were used to transduce healthy murine hematopoietic stem cells (HSCs). On reconstitution of RAG-2-deficient mice with transduced HSCs, the specific vectors were efficiently expressed in T cells, minimally expressed in B cells, and not expressed in immature cells of the bone marrow. Addition of the CD4 gene-silencing element in the vector regulatory sequences led to further restriction of eGFP expression into CD4(+) T cells in reconstituted mice and in ex vivo-transduced human T cells. Non-T CD4(+) dendritic and macrophage cells derived from human CD34(+) cells in vitro expressed the transgene of the specific vectors, albeit at lower levels than CD4(+) T cells. Altogether, we have generated lentiviral vectors that allow specific targeting of transgene expression to CD4(+) cells after differentiation of transduced mice HSCs and human mature T cells. Ultimately, these vectors may prove useful for in situ injections for in vivo gene therapy of HIV infection or genetic immunodeficiencies.
Collapse
Affiliation(s)
- Gilles Marodon
- Centre National de la Recherche Scientifique UMR-7087, Biologie et Thérapeutique des Pathologies Immunitaires, Centre d'Etude et de Recherche en Virologie et en Immunologie, Hôpital La Pitié-Sâlpétrière, Paris, France
| | | | | | | | | | | |
Collapse
|
99
|
Laufs S, Buss EC, Zeller WJ, Fruehauf S. Transfer of drug resistance genes in hematopoietic progenitors for chemoprotection: is it still an option? Drug Resist Updat 2003; 6:57-69. [PMID: 12729804 DOI: 10.1016/s1368-7646(03)00002-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/20/2022]
Abstract
For numerous malignancies a relationship between the intensity of antineoplastic chemotherapy and tumor response has been demonstrated. Myelotoxicity is the main cause of chemotherapy-associated morbidity and of treatment delays. The concept of myeloprotective cytostatic drug resistance gene transfer to normal hematopoietic stem cells (HSC) therefore sparks great enthusiasm. While initial studies using murine retroviral vectors on murine HSC showed that the concept works, a number of clinical studies in the last decade were not informative because of limitations in transduction efficiency and transgene expression.Furthermore, possible side effects such as unforeseen transgene activity and vector integration-based leukemogenesis have been reported. Among others, these developments raised some scepticism against the feasibility of myeloprotective gene transfer. Recently, considerable improvements have been achieved in vector design, HSC manipulation, selection protocols and risk assessment methods which are discussed in detail here. Based on these experimental studies successful clinical trials can now be anticipated.
Collapse
Affiliation(s)
- S Laufs
- Research Program Innovative Cancer Diagnostics and Therapy, German Cancer Research Center, Heidelberg, Germany
| | | | | | | |
Collapse
|
100
|
Abstract
Autoimmune diseases are incurable. We propose a combination curative strategy using haematopoietic stem cells and gene therapy. The strategy should establish molecular chimerism with culprit antigen expressed by donor antigen-presenting cells in the thymus, resulting in immunological tolerance. Our strategy is based on the intrathymic tolerance and disease resistance established by transgenic expression of antigen in the thymus under control of an MHC class II promoter. We propose that transplantation with genetically engineered stem cells provides an advance over current autologous bone marrow transplantation, in that it should remove the significant risk of disease relapse.
Collapse
Affiliation(s)
- Frank Alderuccio
- Department of Pathology and Immunology, Monash University Medical School, Victoria 3181, Australia
| | | | | |
Collapse
|