Transplant Immunology 9 (2002) 301–314
Review
Gene therapy in transplantation D. Chen, R. Sung, J.S. Bromberg* Carl C. Icahn Institute for Gene Therapy and Molecular Medicine and the RecanatiyMiller Transplantation Institute, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1496, New York, NY 10029-6574, USA
Abstract Gene transfer and gene therapy represent a relatively new field that has grown and expanded enormously in the last 5–10 years. The application of gene transfer and gene medicines to transplantation is currently in its infancy. Consideration for gene medicines in transplantation requires delivery of vectors, either to the graft or to the immune system. Delivery of vectors to the graft provides a choice of potential immunologic targets including: costimulatory signals; inhibitory cytokines; adhesion molecules; and molecules relating to apoptosis. In addition, non-immunologic targets, that increase graft protective mechanisms by reducing ischemic and immunologic damage, represent significant targets for gene transfer. Delivery of vectors to the immune system includes potential targets to modify the immune system, and results in tolerance. Other considerations for gene therapy include the development of additional technologies, such as gene conversion or transgenesis coupled with xenotransplantation, which may provide genetically modified organs. Another important aspect of gene transfer relates to regulation of the transgene expression. A variety of issues concerning innate immunity, adaptive immunity, response to vector components, response to transgene products, and entry of vectors into the antigen presentation and processing pathway require further investigation and refinement of approaches. Lastly, regulatable promoters and the understanding of their interaction with individual cells, tissues and organs, and their interaction with innate and adaptive immunity, are of paramount importance to improving the efficacy and utility of gene transfer. There is no doubt that there is much exciting basic and translational science to be accomplished in the next decade in order to solve these potential barriers and advance gene medicines into the clinical realm in transplantation. 䊚 2002 Elsevier Science B.V. All rights reserved. Keywords: Transplantation; Gene therapy; Tolerance
1. Introduction Organ transplantation has become a conventional treatment for failure of various organs, but chronic allograft rejection, the side effects of chronic immunosuppressive therapy and severe donor organ shortages continue to limit its success. Gene therapy has the potential to prevent graft rejection by manipulating the immune response in the microenvironment of the graft, by facilitating the induction of tolerance, or by generating transgenic animals for xenotransplantation. 2. Modification of the graft One of the most important advantages of gene therapy in transplantation is that it can be targeted to a specific *Corresponding author. Tel.: q1-212-241-8938; fax: q1-212-8492437. E-mail address:
[email protected] (J.S. Bromberg).
organ or tissue through ex vivo manipulation. Therefore, local immunosuppression, confined to the transplanted organ or tissue, can be achieved. Almost every transplantable organ or tissue has been shown to be receptive to gene transfer by one or more of the currently available gene transfer vectors. The susceptibility of different organs, tissues, or cells to gene transduction may vary depending on their biological characteristics. For example, when considering the application of gene therapy to tissue transplantation, the cornea stands out as having unique properties. The tissue is small; can be maintained in culture for several weeks at 32–37 8C, instead of a few hours at 0 8C; and the key target cell, the corneal endothelial cell, forms a single sheet of cells on the inner surface of the cornea and is, therefore, perfectly accessible to vectors. Another area of transplantation that is likely to benefit most from advances in gene therapy techniques is that of cell transplantation, in
0966-3274/02/$ - see front matter 䊚 2002 Elsevier Science B.V. All rights reserved. PII: S 0 9 6 6 - 3 2 7 4 Ž 0 2 . 0 0 0 2 3 - 0
302
D. Chen et al. / Transplant Immunology 9 (2002) 301–314
Table 1 Transgenes used in the modification of the graft Transgenes
Mechanism of suppression
Allograft
Reference
CTLA4Ig
Inhibit CD28-CD80y86 interaction, interrupt costimulatory pathway Inhibit cytokine synthesis, negative regulation of antigen presenting cell function Suppress Th1-mediated immune response Block cell adhesion
Rat kidney, liver, islets
w4–6x
Mouse heart, rat heart, sheep cornea
w7–12x
Mouse heart
w13x
Rat heart, mouse islets
w15,16x
Chemokine antagonist
Mouse heart
w14x
Induce apoptosis of immune cells Inhibit inflammatory signals, attenuate ischemia-reperfusion injury Anti-oxidative stress Protect tissue cells from apoptosis
Mouse heart, islets Mouse heart, rat heart
w17,18x w19x
Rat aorta Mouse liver, endothelium Rat heart
w21x w22,23x
vIL-10, IL-10, TGF-b IL-12 p40 ICAM-1 antisense vMIP-II, MC148 FasL NF–kB decoy iNOS Bcl-2 HSP 70
Enhance myocardial tolerance after normothermic ischemia-reperfusion
particular, islet transplantation. Because islets can be placed in culture before transplantation, they have the potential for efficient and targeted transfection. Furthermore, culture allows for selection of transduced cells. Although efficient gene transfer into primary cultures of human or pig vascular endothelial cells has been reported, uptake by vascular endothelial cells in pieces of aorta was found to be much less efficient w1x. Delivery to the myocardium was found to be more efficient than to the artery, with the former resulting in reporter expression in a localized area of the heart which peaked between days 3 and 6 after injection and returned to control levels within a month w2x. As an alternative to directly modifying the graft, genetically engineered cells have been co-transplanted with cellular grafts (for example islets) to produce locally active immunosuppressive bioreagents w3x. Many different biologically active molecules have been delivered into grafts to prolong survival, as outlined in Table 1. Two major approaches can be defined: (1) inhibiting anti-graft immune responses; and (2) increasing graft protective mechanisms against the host immune response. 2.1. Inhibition of anti-graft immune responses 2.1.1. Blockade of costimulatory signals Costimulatory signaling between CD28-CD80y86, or CD40-CD40L, plays a pivotal role in alloimmune responses and transplant allograft rejection. Failure of this receptor–ligand engagement at the time of antigen recognition induces a state of anergy. Administration of anti-CD40L antibody or CTLA4Ig effectively prolongs allograft survival in heart, kidney, liver and islet cell transplantation in rodents and subhuman primates. Gene
w20x
transfer to modify the grafts has been described for CTLA4Ig. Adenovirus-mediated CTLA4Ig gene therapy has resulted in prolonged survival in rat liver, kidney and islet transplantation w4–6x. 2.1.2. Inhibitory cytokines Expression of type 2 cytokines or TGFb has the potential to promote graft survival by inhibiting the production of type 1 cytokines and blocking some antigen presenting cell and macrophage functions. Viral IL-10 (vIL-10) is a viral form of interleukin-10 (IL-10) that is encoded by the Epstein–Barr virus, but lacks some of the T cell immunostimulatory activities of IL10. Gene transfer of the sequence encoding vIL-10 or TGFb have resulted in prolonged allograft survival in various models w7–9x. Cellular IL-10 has also been reported to promote permanent liver graft acceptance w10x and prolong cardiac allograft survival in rats w11x. A recent study reported that in sheep, ex vivo transfer of the gene encoding IL-10 to cornea can prevent rejection after allotransplantation. Five of nine genetically manipulated corneas showed prolonged survival, and another two showed indefinite survival, without any other treatment w12x. Retrovirus-mediated local production of the p40 subunit of IL-12 suppressed Th1mediated immune responses and prevented allogeneic myoblast rejection in the mouse w13x. 2.1.3. Inhibition of leukocyte infiltration or adhesion The recruitment and activation of recipient leukocytes in response to a transplanted organ constitute the initial steps of a complex immune response, which ultimately culminates in rejection of the graft. Chemokines are the primary molecules involved in the leukocyte recruitment. Plasmid-mediated gene transfer of virally encoded
D. Chen et al. / Transplant Immunology 9 (2002) 301–314
chemokine antagonists vMIP-ll and MC148 have been reported can block donor-specific lymphocyte immunity within cardiac allografts and prolong graft survival w14x. Adhesion molecule interactions including LFA-1y ICAM-1, LFA-3-CD2 and VLA-4yVCAM-1 promote interactions between leukocytes and other cells. Transfection of rat hearts with antisense oligonucleotides, for blockade of ICAM-1, demonstrated specific reduction of chronic graft vascular disease w15x. 2.1.4. Inducing apoptosis of immune cells Theoretically, expression of FasL by grafted tissue may protect allogeneic grafts against immune cell infiltration and destruction by inducing apoptosis of graftreactive T cells expressing Fas. But, contradictory results have been obtained. The survival of islet allografts cotransplanted with myoblasts overexpressing FasL was significantly prolonged w17x, whereas mice transplanted with FasL expressing islet allografts were rejected in an accelerated fashion due to massive granulocyte infiltration w18x. These results indicate that regulation of inflammation and immune response by Fas and FasL is complex. 2.2. Increasing graft protective mechanisms It has been postulated that chronic allograft dysfunction is mediated by both alloantigen-dependent (MHC incompatibility, immune mechanism) and alloantigenindependent factors, such as anoxia and ischemia-reperfusion injury that result in inflammatory reactions. Some molecules which have protective actions in non-immune processes, may also help graft survival. 2.2.1. Preventing endothelial activation NF–kB, is an inducible activator of transcription of a large array of genes, some of which are required for inflammatory and immune response. Inhibition of NF– kB may block the activation of endothelial cells, and subsequently protect the graft from inflammatory injury. In one such approach double-stranded oligodeoxynucleotides with a specific affinity for NF–kB (NF–kB decoy), were transfected into rat hearts. Gene transfection of the NF–kB decoy attenuated ischemia-reperfusion injury after prolonged heart preservation w19x. Heat shock protein 70 (HSP70) gene transfection has been shown to enhance myocardial tolerance after normothermic ischemia-reperfusion. In a clinically relevant donor heart preservation protocol, HSP70 gene transfection protects both mechanical and endothelial function w20x. 2.2.2. Anti-oxidative stress Reactive oxygen species are responsible for cell death within the graft, induce cytokine production and recruit leukocytes. Nitric oxide has been shown to suppress T
303
cell proliferation, inhibit leukocyte chemotaxis and prevent vascular smooth muscle cell proliferation. Transduction of rat heart allografts with an inducible nitric oxide synthase gene suppressed the development of allograft arteriosclerosis for 4 weeks w21x. 2.2.3. Anti-apoptotic genes In vitro expression of anti-apoptotic genes (Bcl-2, Bcl-xL and A20) by endothelial cells has been shown not only to protect hepatocyte or endothelial cells from apoptosis (as a consequence of ischemia-reperfusion injury), but also to inhibit their activation through an NF–kB-dependent mechanism w22,23x. 2.2.4. Immunomodulation using dendritic cells Dendritic cells (DC), highly specialized antigen-presenting cells (APC), are now regarded not only as the initiators and regulators of immune responses, but also as potentially powerful tools for the therapeutic manipulation of immune reactivity in allograft rejection as well as cancer, autoimmunity and infectious disease w24x. A role for DC in tolerance induction was recognized initially in the context of intra-thymic self-tolerance. In addition to having a role in central tolerance induction, DC are now regarded as potential modulators of peripheral immune responses w25x. Compared with strategies that aim at directly manipulating the transplanted organ, the induction of T-cell non-reactivity or tolerance via DC manipulation can be fulfilled outside the transplanted tissue and before transplantation. Direct damage to the organ that might be caused by the vectors or immunosuppressive genes used for manipulation could thus be avoided. Another advantage of DC treatment is that they can be manipulated much more easily than solid organs under controlled laboratory conditions. Since DC constitutively express MHC antigens, home to T-dependent areas of recipient lymphoid tissue, and interact selectively with host T cells, they are an attractive target for genetic manipulation to improve the outcome of allograft survival. Studies have reported that immunosuppressive genes such as CTLA4Ig w26x, TGFb w27x and vIL-10 w28x can be successfully delivered into DC and promote T cell hyporesponsiveness. 3. Modification of the host Tolerance remains the primary goal in transplantation immunology. There are several methods that can be used to create a state of acceptance, such as micro- and macrochimerism, co-stimulatory blockade, deletion of donor-specific immune cells, and induction of active, persistent regulatory mechanism w29x. When genetically engineering the host’s immune system, researchers also aim at these mechanisms.
304
D. Chen et al. / Transplant Immunology 9 (2002) 301–314
3.1. DST type protocol—MHC gene transfer and tolerance induction Although MHC molecules are the main targets for allorejection, exposure of the recipient to donor MHC antigens (such as donor blood or bone marrow, MHC molecules, or MHC-derived peptides) in a non-immunogeneic antigen presentation context has been shown to promote graft acceptance. Donor-specific blood transfusion (DST) provided to prospective kidney transplant recipients has resulted in significant improvement in graft survival rates in livingrelated kidney transplant recipients w30x. A state of permanent chimerism may result from the transfusion of stem cells contained in DST w31x, which is considered one of the important tolerance-inducing mechanisms. However, bone marrow may be more effective at establishing chimerism than peripheral blood, because bone marrow has 10–100 times more stem cells than peripheral blood. Indeed, the infusion donor-specific bone marrow (DBM) has been found to be effective in inducing donor-specific graft acceptance w32x. The potential mechanism by which DST and DBT immunomodulate the immune system are likely to be different only in degree rather than kind. These mechanisms include anergy, development of anti-idiotypic antibodies, provision of HLA antigen, alteration in T cell subsets, stimulation of regulatory cells, clonal deletion, regulation of cytokines, andyor chimerism w33x. Among these mechanisms, microchimerism with the concomitant persistence of soluble donor HLA antigen is felt by many to be the most important. With the concern that DST may sensitize patients, while DBM infusion may predispose the recipient to graft-vs.-host disease and is not easily accomplished in living donor organ transplantation, DNA-mediated gene transfer may, represent a safer approach to molecular chimerism. Gene delivery of sequences encoding alloMHC class I antigens to autologous BM cells facilitated the development of tolerance to a mismatched skin graft in mice, by inducing molecular, rather than cellular, chimerism w34x. This strategy might also facilitate the development of unresponsiveness to a fully mismatched vascularized organ, and similar results were reported in a class I transgenic model of heart transplantation w35x. The hyporesponsiveness was likely the result of T cell anergy rather than clonal deletion w36x. Gene delivery of sequences encoding donor MHC class II antigen has also induced prolonged donor-specific organ survival w37x. The high level of MHC polymorphism may not be an obstacle to this approach, since it has been shown that the presentation of a single allelic product can result in inhibition of recipient response to the entire donor haplotype. It is well known that tolerance to self-MHC antigen develops via presentation of this antigen on the thymic
epithelium. Thus, the thymus seems to be a good target for tolerance induction by MHC gene transfer. Delivery of donor cells to the thymus in combination with modification of the peripheral immune system has been shown to induce tolerance to donor tissue subsequently transplanted at peripheral sites w38x. Indirect gene transfer to thymus using myoblasts, myotubes and fibroblasts as cellular intermediates has also been accomplished successfully. This approach, using donor MHC class I presentation to thymus, led to long-term specific tolerance to subsequent liver transplants w39x. Another study used direct gene injection to the thymus to express a single foreign MHC class I Ag in the thymus, prolonging liver allograft survival over 100 days in the majority of recipients following this manipulation w40x. 3.2. Systemic immunosuppression It was reported recently, in a rat lung allograft model, that significant reduction in rejection grade (from grade 3 to 2) occurred after systemic but not intrabronchial adenoviral vector CTLA4Ig gene transduction. The authors concluded that local expression of immunomodulatory proteins could be achieved within lung allograft by intrabronchial delivery of adenoviral vector, but that systemic immunomodulation is required to modify acute rejection w41x. In contrast to protein administration, in vivo gene transfer may be able to produce persistent, biologically controlled long-term expression of gene product. Therefore, gene therapy may have a role in providing long-term systemic immunosuppression. Some studies show that systemically administered adenovirusmediated gene transfer with CTLA4Ig is effective for prolonging both transgene expression and allo- and xenograft survival w42x. Another group reported that a single treatment with AdCD40Ig produced a substantial amount of CD40Ig protein in serum and allowed indefinite graft survival in a stringent rat liver allograft model w43x. These approaches, however, suffer from the same problem of conventional immunosuppression in that chronic systemic administration is associated with a number of adverse events and side effects. Recently, a new type of adenovirus vector, Adex1CALoxCTLA4IgGLox was constructed w44x. It could be manipulated to terminate in vivo CTLA4IgG expression at a desired time point by using Adex1CACre virus. This termination system may minimize the adverse effects of CTLA4Ig gene therapy. It was demonstrated that: CTLA4IgG expression from Adex1CALox-CTL4IgGlox was persistent and efficient; in vivo gene expression was completely terminated after administration of Adex1CACre virus; long-term acceptance of allografts (pancreatic islet and skin) was achieved even after the termination of CTLA4IgG expression; and immune responses against adenovirus were preserved. Thus, the
D. Chen et al. / Transplant Immunology 9 (2002) 301–314
principle of systemic gene transfer and stringent regulation of expression may achieve tolerogenic effects. 3.3. Selective T cell suicide gene therapy Control of anti-donor activated T cells involved in allograft rejection, while preserving immunocompetence, is a challenging goal in transplantation. Engineered T cells expressing a viral thymidine kinase (TK) suicide gene metabolize the non-toxic prodrug ganciclovir (GCV) into a metabolite toxic only to dividing cells. This system is particularly well suited to developing an immunomodulation strategy based on elimination of the dividing T cells engaged in pathological immune responses, while sparing quiescent T cells. In transgenic mice expressing TK in T cells, it has been shown that rejection of fully allogeneic skin graft and non-vascularized heart graft can be significantly delayed by GCV treatment w45x. In addition, a short course of GCV induces long lasting acceptance of vascularized abdominal heart allograft, and the tolerance generated in this system is robust, as shown by the acceptance of a second abdominal allograft, without any additional treatment. These mice remain immunocompetent since they are capable of rejecting third-party allografts with normal kinetics w46x.
305
DNA, and that the retargeted introns retain activity in human cells. This work provides the practical basis for potential applications of targeted group II introns in genetic engineering, functional genomics and gene therapy w48x. 3.5. Gene therapy in xenotransplantation Shortages of human organ for transplantation have made it necessary to examine the possibility of using non-human organs for xenotransplantation. The pig is presently considered the most likely source of organs for human xenotransplantation because it is easy to breed, has compatibly sized organs and offers the possibility of genetic manipulation. The major immediate immunological hurdle that a discordant vascularized xenograft will most probably face is a complementmediated hyperacute rejection (HRA) following the binding of preformed, xenoreactive natural antibodies (XNAs) against xenoantigens. In fact almost all of the human Abs bind to an oligosaccharide pig xenoepitope defined as Gal-a(1,3)-Gal-b(1,4)-GlcNAc-protein, also known as aGal. Attempts to use gene therapy to prevent xenograft rejection also involve the modification of donor organ and the host. 4. Modification of donor organs
3.4. Gene conversion Gene therapy is usually taken to mean the delivery of a gene(s) of interest in an expression vector to a target cell, however, there are alternative approaches to gene therapy, such as targeted DNA or RNA conversion. Although germ line mutations can be incorporated by means of embryonic stem cell technology, little progress has been made toward introducing mutations in somatic cells of living organisms. Triplex-forming oligonucleotides (TFOs) recognize and bind to specific duplex DNA sequences and have been used extensively to modify gene function in cells. It has been reported that TFOs can induce mutations at specific genomic sites in somatic cells of adult mice, providing the initial evidence that sequence-specific, DNA-modifying reagents may prove useful in intact animals w47x. Group II introns are catalytic RNAs that function as mobile genetic elements by inserting directly into target sites in doublestranded DNA, and then reverse-transcribed into genomic DNA by the associated intron-encoded protein. Target site recognition involves modifiable base-pairing interactions between the intron RNA and a )14-nucleotide region of the DNA target site, as well as fixed interactions between the intron-encoded protein and flanking regions. Using human immunodeficiency virus-type 1 (HIV-1) proviral DNA and the human CCR5 gene as examples, it has been shown that group II introns can be retargeted to insert efficiently into virtually any target
Eliminating the aGal epitope from porcine tissue through genetic engineering of pigs would be the most direct way to prevent HAR or delayed xenograft rejection (DXR). It is not yet possible to conduct genetargeting in pigs to knock out the aGal gene because porcine embryonic stem cell lines capable of achieving germ line transmission of mutations induced by homologous recombination are not available. Preventing HAR by genetic transfer of human complement regulators has been another major focus. Several groups have successfully introduced genes that encode human complement regulators, such as human decayaccelerating factor (DAF), membrane cofactor protein (MCP) or CD59, into the pronuclei of fertilized pig oocytes w49x. Organs from DAF-expressing pigs survived for up to 8 days when transplanted into baboons, in the continued absence of any visible HAR w50x. In these studies, there was no attempt to reduce the titers of xenoreactive natural antibodies or to provide other immunosuppression, clearly indicating that HAR can be surmounted by the local inhibition of complement activation by genetically manipulating the donor organ. Both HAR and DXR involve the activation of endothelial cells (ECs), so modification of the graft by transferring protective genes is another important approach to xenograft survival. Transfer of genes encoding CTLA4Ig w51x, IL-10 and TGF-b w52x has been reported to result in significant prolongation of islet
306
D. Chen et al. / Transplant Immunology 9 (2002) 301–314
xenograft survival. The anti-inflammatory properties of hemoxygenase-1 (HO-1) have been shown to promote xenograft survival, and rapid synthesis of HO-1 in cardiac xenografts is essential for ensuring long-term xenograft survival w53x. Another approach is to eliminate or reduce aGal expression by replacing aGal epitopes with other carbohydrate moieties to which humans are immunologically tolerant. This method involves the genetic engineering of xenogenetic cells to express a-(1,2)fucosyltransferase (a-FT), which adds fucose residues to an oligosaccharide. In both pigs and humans, the substrate for the fucosyltransferase is identical. It has been shown that expression of a-FT in vascular endothelium can eliminate approximately 90% of cell surface aGal epitopes w54x, although the remaining 105;106 aGal epitopes is sufficient to permit DXR mediated by XNAs without further immunologic manipulation w55x. 5. Modification of the host Lasting mixed chimerism was achieved in alpha1, 3galactosyltransferase knockout (GalT KO) mice by cotransplantation of GalT KO and GalTqyq wild-type (WT) marrow after lethal irradiation. Mixed chimerism efficiently induces anti-Gal-specific B cell tolerance in addition to T cell tolerance, providing a single approach to overcoming both the humoral and the cellular immune barriers to discordant xenotransplantation w56x. Currently, this approach is not yet practical in humans. In another report, a functioning gene that encodes agalactosyltransferase (aGT) was introduced by retroviral gene transfer into autologous murine bone marrow cells, preventing aGal XNA production w57x. Thus, genetic engineering of bone marrow may overcome humoral rejection of discordant xenografts and may be useful for inducing B cell tolerance. MHC gene transfer into BM cells have also shown promising results in controlling cellular responses to xenoantigens. Transfer and expression of xenogeneic MHC class II DR transgenes can be achieved in baboons. This therapy may prevent late T cell-dependent responses to porcine xenografts, which include induced non-aGal IgG antibody responses w58x. 6. Limitations to gene transfer and expression A variety of barriers exist to efficient, targeted expression by gene therapy vectors. To achieve this goal, vectors must selectively reach and be retained by the target tissue, enter cells and negotiate endosomal compartments. Vector-derived nucleic acid must be transported to the nucleus, and must be efficiently transcribed. At each of these levels, conditions particular to the vector and the intended application may ultimately impair effective expression of transgene. In addition, host innate and adaptive immune responses to vector
components and transgene products limit vector expression through apoptosis, T-cell mediated cytotoxicity, vector clearance and inhibition of transcription. 6.1. Adaptive immune responses to vector antigens Adenoviral vectors, although highly efficient, are unfortunately also highly immunogenic. These vectors are attractive for gene transfer because of their broad host range, and capacity for large DNA inserts. However, administration of first-generation adenovirus vectors (those with viral genes E1 and E3 deleted) generates well-characterized adaptive immune responses against viral capsid proteins, which appear to be the primary mechanism by which expression is extinguished w59,60x. Both the cellular and humoral arms of the adaptive immune response are triggered, the former effecting viral clearance, and the latter preventing effective readministration of vector. These responses have been attributed to low levels of viral gene expression, or may reflect contamination by wild-type virus. The importance of viral gene expression is supported by a diminution of both Th2 responses and neutralizing antibodies when E4 is deleted from first-generation vectors w61x, and by the absence of late inflammation in muscle when vectors lacking all viral genes are employed w62x. However, cellular responses may not necessarily require viral gene expression, as vectors rendered biologically inactive generate similar infiltration of CD4q and CD8q lymphocytes as do untreated vectors, and antivector CTL responses can be generated in the absence of vector transcription w63,64x. Adeno-associated virus (AAV) vectors, although less efficient than adenovectors, incite much less of an immune response. Consequently, expression by these vectors results in greater vector persistence and transgene expression w65x. Transfection of dendritic cells (DC) by adenovirus vectors is an important component in the development of the anti-vector immune response. Adenovirus vectors efficiently transfect both mature and immature DC without altering maturation or function w66x. DC transfected by adenovirus can be adoptively transferred to effect immune mediated elimination of adenovirus or AAV infected cells, but AAV-transfected DCs cannot. This is likely due to the inability of AAV to transduce or to effectively activate DCs, and may account for the decreased immunogenicity of AAV vectors w67,68x. Dendritic cells infected in vitro with adenovirus encoding an antigenic peptide can subsequently induce strong specific CTL responses that can be augmented with repeated administration of transfected DC w69x. Low titers of neutralizing antibody are generated with this method; in contrast, direct immunization generates high titers of neutralizing antibody, which limits the efficacy of repeated administration. Such an approach may have
D. Chen et al. / Transplant Immunology 9 (2002) 301–314
advantages in designing vaccination therapies or in tumor immunization. 6.2. Adaptive immune responses to transgenes Immune responses to encoded transgenes also limit expression by gene therapy vectors. Much of the literature describing immunity to adenovirus vectors has been generated using vectors expressing the immunogenic reporter b–galactosidase. Human a1-antitrypsin and human factor IX are two examples of less immunogenic reporter proteins whose duration of expression by adenovirus vectors exceeds that of b–galactosidase w70,71x. Long-term expression has also been demonstrated in mice transgenic for hAAT and b–galactosidase w70,72x. Vectors encoding murine erythropoietin demonstrate superior long-term expression in mice compared with identical vectors encoding the human homologue w73x. Thus, while immune responses to adenovirus proteins per se may be less important in limiting expression than originally believed, the immune response to the vector may potentiate the antitransgene response; immune responses to Factor IX, while activated by adenoviral vectors, are absent following administration of AAV vectors expressing Factor IX w74x. The magnitude of immune responses to transgene products may be more strain-dependent than responses to viral antigens, which may account for the strain variation in expression by adenoviral vectors w75x. Regardless of the relative contributions of responses to vector antigens and transgene products, it is clear that any gene therapy strategy must account for both in order to achieve long-term expression. The mechanisms underlying adaptive immune responses to transgenes also include a prominent role for DC not only in indirect presentation of soluble antigen, but also directly via vector uptake. Inoculation of plasmid DNA into muscle or skin results in activation of plasmid-containing dendritic cells at the injection site, in draining lymph nodes and in the systemic circulation w76,77x. These cells function as antigen presenting cells (APC), can induce specific proliferation of CD4q T cells and generation of CD8q CTLs, and can induce both primary and secondary adaptive immune responses w78,79x. Much of the transgene product can be presented directly by transfected DC, since antigen-bearing DC in draining lymph nodes can be depleted by antibodies to a cell surface protein encoded by cotransfected DNA w79x. This direct pathway appears to be the primary mode of antigen presentation in this model, rather than cross- or indirect presentation of antigen by untransfected DC that have taken up soluble antigen. 6.3. Immune responses to nucleic acids Immune responses to nucleic acids in gene therapy vectors also contribute to limitation of expression. Bac-
307
terial DNA is a powerful stimulator of polyclonal proliferation in B cells and cytokine production by monocytes and lymphocytes w80,81x The immunostimulatory properties of bacterial DNA are attributed to unmethylated CpG motifs, which may be present in plasmid DNA or synthetic oligonucleotides. These motifs are much less frequent in mammalian DNA, which is not immunostimulatory w82x. These responses are sensitive to inhibitors of endosomal acidification such as bafilomycin A or chloroquine w83,84x. This acidification is coupled to the generation of intracellular reactive oxygen species, which in turn is linked to the degradation of IkB w83x, and inhibition of IkB degradation suppresses the immunostimulatory effects of CpG DNA w85x. CpG DNA also induces the activation of cJun N-terminal kinase and p38 which leads ultimately to activation of the transcription factor AP-1 w86,87x. The actions of CpG motifs stimulate innate immune responses via TNFa and IL-6 secretion, monocyte activation and increased NK cell activity w83,88,89x. In addition, type I interferons, IFNg, IL-12 and IL-18 are also induced, and subsequently stimulate Th1 responses and enhance adaptive immunity w90–93x. Although CpG motifs are generally immunostimulatory to most cell types, evidence exists that different sequences possess different cellular profiles of activity w82x. While the ability of these motifs to function as immune adjuvants to coadministered antigens are advantageous for immunization, they are potentially deleterious to gene therapy applications, which require sustained expression of transgene product. Efforts to minimize CpG immunostimulation by methylation of stimulatory CpG sequences, while effective in reducing cytokine induction, have resulted in dramatic decreases in gene expression due to methylation of CpG sequences in the promoter w82x. 6.4. Viral components Vector efficiency is also limited by the ability to enter host cells via defined receptors. The tissue distribution of cellular receptors for viral vectors may influence the distribution of vector and can result in either inefficient transduction of target tissue or in transduction of undesirable cell types. Adenoviral vector infection can be mediated by the cocksackie and adenovirus receptor (CAR), av-integrins or MHC class I heavy chain w94x. Tissue distribution of CAR expression varies in a fashion that is similar among species w95x. CAR expression is greatest in liver, which is also the organ with the greatest transgene expression following systemic administration. More efficient vector transduction and transgene expression has been achieved in typically inaccessible tissues such as lung and brain in CAR transgenic mice, which express higher levels of CAR on these tissues, compared with wild-type mice w96x. Enhanced expression and the ability to express transgene following repetitive admin-
308
D. Chen et al. / Transplant Immunology 9 (2002) 301–314
istration of vector has also been reported with adenoviral vectors that encode the CAR receptor in addition to reporter gene w97x. In this model, enhanced CAR expression on the target tissue presumably permits greater cell entry of vector following secondary administration. However, despite the important role of CAR binding, other barriers to effective expression exist. Many tissues expressing levels of CAR and av –integrins, which would make them potential targets for adenoviral vectors, do not express well in vivo; indeed, receptor expression and in vivo expression patterns correlate poorly w95x. Presumably significant anatomic barriers to vector accessibility to target tissues exist. Mutations of the adenoviral knob domain of the fiber protein, which binds CAR, do not alter the in vivo biodistribution of vector compared to wild-type vector w98x. However, replacing the CAR-binding domain with the integrinbinding motif RGD results in an expanded vector tropism in vivo w99x. Other barriers to efficient, targeted adenovirus infection include endothelial cells of solid organs, antiadherence layers of glycosaminoglycans in urothelium and diffusion of vector to distant sites in applications where direct injection of vector is employed. Targeting of adenoviral vectors to tissues such as endothelial cells which have low transduction efficiency has been achieved by utilization of recombinant bispecific antibodies which serve as a bridge between the knob domain and tissue-specific cell surface proteins w100x. Administering vectors in specific polyamide vehicles, which abrogate the GAG barrier w101x, has enhanced transfection to urothelial cell types such as bladder. Enhanced local expression of vector and decreased peripheral transduction of non-target tissues has been achieved by complexing adenoviral vectors in a collagen-avidin gel using a specific anti-adenoviral IgG w102x. Expression by retroviral vectors is also restricted by tissue distribution of cell surface receptors. Retroviruses such as amphotrophic murine leukemia virus (MuLV) and gibbon ape leukemia virus (GALV) possess a broad tissue tropism due to the ubiquitous expression of their cell surface receptors, the phosphate transporter molecules Pit1 and Pit2, respectively. Pseudotyping of other retroviral vectors such as feline leukemia virus with chimeric envelope proteins has produced vectors capable of binding either of these receptors, thereby enhancing the potential infectivity of these vectors w103x. Envelope pseudotyping may also effect tissue selectivity of retroviral vectors; vectors have been successfully pseudotyped to target melanoma cells w104x and human lymphocytes w105x. However, others directed against a folate receptor, EGF receptor and IGF receptor have resulted in impairment of virus transduction, implying that other barriers to vector entry may exist w106–108x.
6.5. Antigen processingypresentation pathways By their nature gene transfer vectors must effect the transfer of nucleic acid through endosomal compartments, and cytoplasm to the nucleus for transcriptional activation. The type of gene transfer vehicle influences the rate of transfer of DNA to acidic lysosomes, the degree of intracellular recycling and exocytosis of DNA, and thus influence expression w109x. These vectors are invariably introduced into antigen processing and presentation pathways, which engender antivector immune responses. Antigen presentation may ensue due to direct transduction of APC by vectors, or by transfer of proteins from target tissue to APC w110–113x. Both viral and non-viral vectors can efficiently direct antigen to MHC class I and class II pathways w114–116x. Retroviral nucleic acid sequences have also been shown to upregulate MHC class I expression in target tissue w117x. Other investigators have shown that double stranded polynucleotides in the cytoplasm of target cells perturb MHC expression and increase the expression of proteasome proteins, antigen peptide transporters and costimulatory molecules, thereby enhancing antigen presentation and recognition w118x. Viral vector antigens also interfere with antigen presentation pathways through changes in the expression or function of MHC molecules. Adenovirus E1 region proteins decrease transcriptional activation of class I genes w119x. The adenovirus E3-19K product binds to class I MHC in the endoplasmic reticulum, resulting in decreased cell surface expression of class I MHC and decrease cytotoxic T cell responses to adenoviral antigens w120–123x. Herpesviruses express genes which selectively interfere with viral antigen presentation by MHC class I, and retroviral vectors encoding HSV ICP47, HCMV US3, or HCMV US11 proteins all decrease target tissue MHC class I expression and evade CTL recognition w124x. These virally encoded proteins may be potentially utilized to potentiate vector persistence and expression. 6.6. Innate immunity While the major mechanism for the attenuation of gene expression is the specific cellular immune response mediated by cytotoxic T lymphocytes and immunoglobulins to either viral proteins and to the encoded transgene product, loss of transgene expression frequently precedes the development of specific immune responses. Thus, non-specific, innate immune mechanisms must also contribute to limiting vector efficiency and persistence w125,126x. Resident macrophages clear a vast majority of adenovirus vector genomes within 24 h of administration w127,128x, and Kupffer cell depletion by gadolinium chloride treatment substantially reduces this vector clearance in the liver w129x. Innate immune responses
D. Chen et al. / Transplant Immunology 9 (2002) 301–314
are characterized by pattern recognition receptors such as mannan-binding lectin, which focus on highly conserved sequences common to many pathogens. Effector responses are rapid and do not require effector cell proliferation. Signaling receptors, such as those of the toll family, induce expression of a variety of cytokines and chemokines involved in innate and adaptive immune responses w130x. A broad range of cytokines (TNFa, IL-6, IFNg, IL-1b) and chemokines (MCP-1, RANTES, IP-10, MIP-1a) are induced following vector administration. While some of these cytokines such as TNFa and IFNg possess intrinsic antiviral properties, they also induce a profound inflammatory response mediated by neutrophils, NK cells and monocytes, which effect vector clearance and apoptosis of transduced cells w125,126,129,131x. In addition, so-called ‘natural interferon producing cells’, recently shown to be dendritic cell precursors, are the chief producers of IFNa and – b upon viral challenge, and are critical components of the antiviral immune response w132–134x. While innate immunity to gene therapy vectors has primarily been characterized in response to viral vectors in mice, similar cytokine and inflammatory responses have also been demonstrated to non-viral vectors w135x, and to adenoviral vectors in non-human primates w136x. As with anti-vector adaptive immunity, this inflammatory response is often not dependent on viral or transgene expression, as such responses occur very early or immediately after infection, and adenoviral vectors progressively deleted of viral genes incite similar inflammatory responses and interference with vector persistence w137,138x. Furthermore, specific antigen recognition may not be required w139x, suggesting that infection of parenchymal cells, that are not part of the leukocytic immune system, induces an innate stereotyped response in these cells that inhibit vector function and gene expression w140–142x. This may occur by apoptosis of vector-transduced cells or by inhibition of vector promoter expression by cytokines w143x. The cytokines induced following virus or viral vector administration are potent inducers of proinflammatory chemokines such as RANTES, MCP-1, MIP-1a, MIP1b, and MIP-2 w131,137,144,145x. Specific inhibition of TNFa or MIP-2 can attenuate virus-induced inflammation w137,146x. Many of these effects are mediated through NF–kB activation, as inhibition of NF–kB activation either by overexpression of its inhibitor IkB or by expression of a mutant of the p65yRelA subunit of NF–kB leads to inhibition of cytokine expression, chemokine expression, and leukocyte infiltration w145,147,148x. However, NF–kB also appears to exert a protective effect on vector-induced hepatic injury; NF–kB inhibition by introduction of an expression vector encoding IkB results in greater susceptibility to TNFa-mediated apoptosis, although antagonism of p65y RelA binding does not. Such protection may be due to
309
feedback inhibition of TNFa production by the p50 subunit of NF–kB w149x. Vector persistence during NF– kB inhibition by IkB overexpression is enhanced only by simultaneous overexpression of the anti-apoptotic gene bcl-2 w147x. 6.7. Promoter extinctionyattenuation The type of promoter directing transgene expression influences expression by gene transfer vectors. A wide variety of promoter types have been studied, and the most commonly used are viral promoters such as MMLV, RSV, SV40, or CMV, but cellular promoters such as the b–actin promoter have also been utilized. Other promoter characteristics may have dramatic influences on transgene expression. The efficiency of the CMVie promoter differs depending on whether the promoter is derived from human or murine CMV, on the size of the promoter fragment used in the vector, and on the cell type transfected in vitro w150x. The nature of the promoter may determine interactions with a number of transcription factors or viral gene products, which may ultimately determine vector efficacy. For example, the adenoviral E4 region gene products can activate gene expression in cis or in trans directed by an RSV but not a CMV promoter w151x. Interactions of promoters with components of the immune system represent an important means by which transgene expression is attenuated. Interferons and TNFa are known to have a variety of antiviral activities. Interferon–g regulates MCMV infection and inhibits reactivation of MCMV from latency w152x. Such inhibition may occur by blockade of replication w153x, but there is evidence that interferons inhibit the onset of MCMVie gene transcription as well, implying a more direct effect on promoter function w141,154,155x. Inhibition of transgene expression by interferons at a posttranscriptional level has been demonstrated in HBV infection, and in MMLV-based retroviral vectors w156,157x. In contrast, CMV promoter activity, which increases following adenovirus vector administration, is stimulated by the transcription factor NF–kB w158x. This may explain the early efficiency of CMV promoter based vectors, as NF–kB activation can be induced by TNFa, IL-1 and IL-6, all of which are stimulated by vector administration. Cytokines induced by immune responses to gene transfer vectors may also exert competitive or synergistic effects on transgene expression at the promoter level. We have shown that IFNg and TNFa in combination are synergistic in inhibition of b–gal expression by adenoviral, retroviral and plasmid vectors in vitro, and by adenoviral vectors in vivo w141x. In contrast, NF–kB-dependent HIV gene expression is stimulated by TNFa but inhibited by IFNa by competitive binding of transcription factors induced by these cytokines to the NF–kB coactivator p300 w159x. Wheth-
310
D. Chen et al. / Transplant Immunology 9 (2002) 301–314
er an individual cytokine inhibits or augments transgene expression may also be determined by vector promoter structure; for example, the deletion or mutation of a transcription factor binding site may lead to loss of inhibition by a given cytokine. Precise characterization of the interactions between cytokines and promoter elements may ultimately lead to gene therapy vectors with promoters designed to capitalize on the cytokine milieu of the target tissue, resulting in augmentation of expression rather than attenuation. References w1x Merrick AF, Shewring LD, Sawyer GJ, Gustafsson KT, Fabre JW. Comparison of adenovirus gene transfer to vascular endothelial cells in cell culture, organ culture and in vivo. Transplantation 1996;62(8):1085 –1089. w2x Muhlhauser J, Jones M, Yamada I, et al. Safety and efficacy of in vivo gene transfer into the porcine heart with replicationdeficient, recombinant adenovirus vectors. Gene Ther 1996;3(2):145 –153. w3x Chahine AA, Yu M, McKernan MM, Stoeckert C, Lau HT. Immunomodulation of pancreatic islet allografts in mice with CTLA4Ig secreting muscle cells. Transplantation 1995;59(9):1313 –1318. w4x Olthoff KM, Judge TA, Gelman AE, et al. Adenovirusmediated gene transfer into cold-preserved liver allografts: survival pattern and unresponsiveness following transduction with CTLA4Ig. Nat Med 1998;4(2):194 –200. w5x Feng S, Quickel RR, Hollister-Lock J, et al. Prolonged xenograft survival of islets infected with small doses of adenovirus expressing CTLA4Ig. Transplantation 1999;67(12):1607 – 1613. w6x Tomasoni S, Azzollini N, Casiraghi F, Capogrossi MC, Remuzzi G, Benigni A. CTLA4Ig gene transfer prolongs survival and induces donor-specific tolerance in a rat renal allograft. J Am Soc Nephrol 2000;11(4):747 –752. w7x Qin L, Chavin KD, Ding Y, et al. Multiple vectors effectively achieve gene transfer in a murine cardiac transplantation model. Immunosuppression with TGF-beta 1 or vIL-10. Transplantation 1995;59(6):809 –816. w8x Qin L, Chavin KD, Ding Y, et al. Retrovirus-mediated transfer of viral IL-10 gene prolongs murine cardiac allograft survival. J Immunol 1996;156(6):2316 –2323. w9x Brauner R, Nonoyama M, Laks H, et al. Intracoronary adenovirus-mediated transfer of immunosuppressive cytokine genes prolongs allograft survival. J Thorac Cardiovasc Surg 1997;114(6):923 –933. w10x Shinozaki K, Yahata H, Tanji H, Sakaguchi T, Ito H, Dohi K. Allograft transduction of IL-10 prolongs survival following orthotopic liver transplantation. Gene Ther 1999;6(5):816 – 822. w11x David A, Chetritt J, Guillot C, et al. Interleukin-10 produced by recombinant adenovirus prolongs survival of cardiac allografts in rats. Gene Ther 2000;7(6):505 –510. w12x Klebe S, Sykes P, Coster D, Krishnan And R, Williams K. Prolongation of sheep corneal allograft survival by ex vivo transfer of the gene encoding interleukin-10. Transplantation 2001;71(9):1207 –1209. w13x Kato K, Shimozato O, Hoshi K, et al. Local production of the p40 subunit of interleukin 12 suppresses T-helper 1-mediated immune responses and prevents allogeneic myoblast rejection. Proc Natl Acad Sci USA 1996;93(17):9085 –9089.
w14x DeBruyne LA, Li K, Bishop DK, Bromberg JS. Gene transfer of virally encoded chemokine antagonists vMIP-II and MC148 prolongs cardiac allograft survival and inhibits donor-specific immunity. Gene Ther 2000;7(7):575 –582. w15x Poston RS, Ennen M, Pollard J, Hoyt EG, Billingham ME, Robbins RC. Ex vivo gene therapy prevents chronic graft vascular disease in cardiac allografts. J Thorac Cardiovasc Surg 1998;116(3):386 –396. w16x Katz SM, Bennett F, Stecker K, et al. ICAM-1 antisense oligodeoxynucleotide improves islet allograft survival and function. Cell Transplant 2000;9(6):817 –828. w17x Lau HT, Yu M, Fontana A, Stoeckert CJ. Prevention of islet allograft rejection with engineered myoblasts expressing FasL in mice. Science 1996;273(5271):109 –112. w18x Kang SM, Schneider DB, Lin Z, et al. Fas ligand expression in islets of Langerhans does not confer immune privilege and instead targets them for rapid destruction. Nat Med 1997;3(7):738 –743. w19x Sakaguchi T, Sawa Y, Fukushima N, et al. A novel strategy of decoy transfection against nuclear factor-kappaB in myocardial preservation. Ann Thorac Surg 2001;71(2):624 –629. w20x Jayakumar J, Suzuki K, Khan M, et al. Gene therapy for myocardial protection: transfection of donor hearts with heat shock protein 70 gene protects cardiac function against ischemia-reperfusion injury. Circulation 2000;102(19 Suppl 3):III302 –306. w21x Shears LL, Kawaharada N, Tzeng E, et al. Inducible nitric oxide synthase suppresses the development of allograft arteriosclerosis. J Clin Invest 1997;100(8):2035 –2042. w22x Bilbao G, Contreras JL, Gomez-Navarro J, et al. Genetic modification of liver grafts with an adenoviral vector encoding the Bcl-2 gene improves organ preservation. Transplantation 1999;67(6):775 –783. w23x Badrichani AZ, Stroka DM, Bilbao G, Curiel DT, Bach FH, Ferran C. Bcl-2 and Bcl-XL serve an anti-inflammatory function in endothelial cells through inhibition of NF-kappaB. J Clin Invest 1999;103(4):543 –553. w24x Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature 1998;392(6673):245 –252. w25x Steptoe RJ, Thomson AW. Dendritic cells and tolerance induction. Clin Exp Immunol 1996;105(3):397 –402. w26x Lu L, Gambotto A, Lee WC, et al. Adenoviral delivery of CTLA4Ig into myeloid dendritic cells promotes their in vitro tolerogenicity and survival in allogeneic recipients. Gene Ther 1999;6(4):554 –563. w27x Lee WC, Zhong C, Qian S, et al. Phenotype, function and in vivo migration and survival of allogeneic dendritic cell progenitors genetically engineered to express TGF-beta. Transplantation 1998;66(12):1810 –1817. w28x Takayama T, Nishioka Y, Lu L, Lotze MT, Tahara H, Thomson AW. Retroviral delivery of viral interleukin-10 into myeloid dendritic cells markedly inhibits their allostimulatory activity and promotes the induction of T-cell hyporesponsiveness. Transplantation 1998;66(12):1567 –1574. w29x Bromberg JS, Murphy B. Routes to allograft survival. J Clin Invest 2001;107(7):797 –798. w30x Salvatierra O, Vincenti F, Amend W, et al. Deliberate donor specific blood transfusions prior to living renal transplantation: a new approach. Ann Surg 1980;192:543 –552. w31x de Waal LP, van Twuyver E. Blood transfusion and allograft survival: is mixed chimerism the solution for tolerance induction in clinical transplantation? Crit Rev Immunol 1991;10(5):417 –425. w32x Sykes M, Sachs DH. Bone marrow transplantation as a means of inducing tolerance. Semin Immunol 1990;2:401 –417.
D. Chen et al. / Transplant Immunology 9 (2002) 301–314 w33x Brennan DC, Mohanakumar T, Flye MW. Donor-specific transfusion and donor bone marrow infusion in renal transplantation tolerance: a review of efficacy and mechanisms. Am J Kidney Dis 1995;26(5):701 –715. w34x Sykes M, Sachs DH, Nienhuis AW, Pearson DA, Moulton AD, Bodine DM. Specific prolongation of skin graft survival following retroviral transduction of bone marrow with an allogeneic major histocompatibility complex gene. Transplantation 1993;55(1):197 –202. w35x Fraser CC, Sykes M, Lee RS, Sachs DH, LeGuern C. Specific unresponsiveness to a retrovirally-transferred class I antigen is controlled through the helper pathway. J Immunol 1995;154(4):1587 –1595. w36x Wong W, Stranford SA, Morris PJ, Wood KJ. Retroviral gene transfer of a donor class I MHC gene to recipient bone marrow cells induces tolerance to alloantigens in vivo. Transplant Proc 1997;29(1–2):1130. w37x Emery DW, Sablinski T, Shimada H, et al. Expression of an allogeneic MHC DRB transgene, through retroviral transduction of bone marrow, induces specific reduction of alloreactivity. Transplantation 1997;64(10):1414 –1423. w38x Posselt AM, Barker CF, Tomaszewski JE, Markmann JF, Choti MA, Naji A. Induction of donor-specific unresponsiveness by intrathymic islet transplantation. Science 1990;249(4974): 1293 –1295. w39x Knechtle SJ, Wang J, Jiao S, Geissler EK, Sumimoto R, Wolff J. Induction of specific tolerance by intrathymic injection of recipient muscle cells transfected with donor class I major histocompatibility complex. Transplantation 1994;57(7):990 – 996. w40x Knechtle SJ, Wang J, Graeb C, et al. Direct MHC class I complementary DNA transfer to thymus induces donor-specific unresponsiveness, which involves multiple immunologic mechanisms. J Immunol 1997;159(1):152 –158. w41x Ugurlu MM, Griffin MD, O’Brien T, Tazelaar HD, Miller VM, McGregor CG. The effects of CTLA-4Ig on acute lung allograft rejection: a comparison of intrabronchial gene therapy with systemic administration of protein. Transplantation 2001;71(12):1867 –1871. w42x Hayashi S, Namii Y, Guan-lin M, et al. Effect of adenovirusmediated gene transfer with CTLA4-Ig gene in organ transplantation. Transplant Proc 1999;31:1944 –1945. w43x Nomura M, Yamashita K, Murakami M, et al. Novel CD40IgG adenovirus-mediated gene therapy as a potent immunosuppressive treatment for liver transplantation in rats. Transplant Proc 2001;33(1–2):189. w44x Takehara M, Murakami M, Inobe M, et al. Long-term acceptance of allografts by in vivo gene transfer of regulatable adenovirus vector containing CTLA4IgG and loxP. Hum Gene Ther 2001;12(4):415 –426. w45x Thomas-Vaslin V, Bellier B, Cohen JL, et al. Prolonged allograft survival through conditional and specific ablation of alloreactive T cells expressing a suicide gene. Transplantation 2000;69(10):2154 –2161. w46x Braunberger E, Cohen JL, Boyer O, et al. T-Cell suicide gene therapy for organ transplantation: induction of long-lasting tolerance to allogeneic heart without generalized immunosuppression. Mol Ther 2000;2(6):596 –601. w47x Guo H, Karberg M, Long M, Jones 3rd JP, Sullenger B, Lambowitz AM. Group II introns designed to insert into therapeutically relevant DNA target sites in human cells. Science 2000;289(5478):452 –457. w48x Vasquez KM, Narayanan L, Glazer PM. Specific mutations induced by triplex-forming oligonucleotides in mice. Science 2000;290(5491):530 –533.
311
w49x Fodor WL, Williams BL, Matis LA, et al. Expression of a functional human complement inhibitor in a transgenic pig as a model for the prevention of xenogeneic hyperacute organ rejection. Proc Natl Acad Sci USA 1994;91(23):11153 –11157. w50x McCurry KR, Kooyman DL, Alvarado CG, et al. Human complement regulatory proteins protect swine-to-primate cardiac xenografts from humoral injury. Nat Med 1995;1(5):423 – 427. w51x Guo Z, Mital D, Mo YY, et al. Effect of gene gun-mediated CTLA4IG and Fas ligand gene transfection on concordant xenogeneic islet graft rejection. Transplant Proc 1998;30(2):589. w52x Deng S, Ketchum RJ, Kucher T, et al. IL-10 and TGF-beta gene transfer for xenogeneic islet transplantation: comparison of effect in concordant vs. discordant combination. Transplant Proc 1997;29(4):2204 –2205. w53x Soares MP, Lin Y, Anrather J, et al. Expression of heme oxygenase-1 can determine cardiac xenograft survival. Nat Med 1998;4(9):1073 –1077. w54x Cohney S, McKenzie IF, Patton K, et al. Down-regulation of Gal alpha(1,3)Gal expression by alpha1,2-fucosyltransferase: further characterization of alpha1,2-fucosyltransferase transgenic mice. Transplantation 1997;64(3):495 –500. w55x Galili U, LaTemple DC, Walgenbach AW, Stone KR. Porcine and bovine cartilage transplants in cynomolgus monkey: II. Changes in anti-Gal response during chronic rejection. Transplantation 1997;63(5):646 –651. w56x Yang YG, deGoma E, Ohdan H, et al. Tolerization of antiGalalpha1-3Gal natural antibody-forming B cells by induction of mixed chimerism. J Exp Med 1998;187(8):1335 –1342. w57x Bracy JL, Sachs DH, Iacomini J. Inhibition of xenoreactive natural antibody production by retroviral gene therapy. Science 1998;281(5384):1845 –1847. w58x Ierino FL, Gojo S, Banerjee PT, et al. Transfer of swine major histocompatibility complex class II genes into autologous bone marrow cells of baboons for the induction of tolerance across xenogeneic barriers. Transplantation 1999;67(8):1119 –1128. w59x Yang Y, Ertl HC, Wilson JM. MHC class I-restricted cytotoxic T lymphocytes to viral antigens destroy hepatocytes in mice infected with E1-deleted recombinant adenoviruses. Immunity 1994;1(5):433 –442. w60x Yang Y, Nunes FA, Berencsi K, Furth EE, Gonczol E, Wilson JM. Cellular immunity to viral antigens limits E1-deleted adenoviruses for gene therapy. Proc Natl Acad Sci USA 1994;91(10):4407 –4411. w61x Chirmule N, Hughes JV, Gao GP, Raper SE, Wilson JM. Role of E4 in eliciting CD4 T-cell and B-cell responses to adenovirus vectors delivered to murine and nonhuman primate lungs. J Virol 1998;72(7):6138 –6145. w62x Chen HH, Mack LM, Choi SY, Ontell M, Kochanek S, Clemens PR. DNA from both high-capacity and first-generation adenoviral vectors remains intact in skeletal muscle. Hum Gene Ther 1999;10(3):365 –373. w63x Kafri T, Morgan D, Krahl T, Sarvetnick N, Sherman L, Verma I. Cellular immune response to adenoviral vector infected cells does not require de novo viral gene expression: implications for gene therapy. Proc Natl Acad Sci USA 1998;95(19):11377 –11382. w64x Molinier-Frenkel V, Gahery-Segard H, Mehtali M, et al. Immune response to recombinant adenovirus in humans: capsid components from viral input are targets for vector-specific cytotoxic T lymphocytes. J Virol 2000;74(16):7678 –7682. w65x Fisher KJ, Jooss K, Alston J, et al. Recombinant adenoassociated virus for muscle directed gene therapy. Nat Med 1997;3(3):306 –312.
312
D. Chen et al. / Transplant Immunology 9 (2002) 301–314
w66x Zhong L, Granelli-Piperno A, Choi Y, Steinman RM. Recombinant adenovirus is an efficient and non-perturbing genetic vector for human dendritic cells. Eur J Immun 1999;29(3):964 –972. w67x Lieber A, He CY, Meuse L, Himeda C, Wilson C, Kay MA. Inhibition of NF-kappaB activation in combination with bcl-2 expression allows for persistence of first-generation adenovirus vectors in the mouse liver. J Virol 1998;72(11):9267 –9277. w68x Jooss K, Yang Y, Fisher KJ, Wilson JM. Transduction of dendritic cells by DNA viral vectors directs the immune response to transgene products in muscle fibers. J Virol 1998;72(5):4212 –4223. w69x Brossart P, Goldrath AW, Butz EA, Martin S, Bevan MJ. Virusmediated delivery of antigenic epitopes into dendritic cells as a means to induce CTL. J Immunol 1997;158(7):3270 –3276. w70x Wadsworth SC, Zhou H, Smith AE, Kaplan JM. Adenovirus vector-infected cells can escape adenovirus antigen-specific cytotoxic T-lymphocyte killing in vivo. J Virol 1997;71(7):5189 –5196. w71x Poller W, Schneider-Rasp S, Liebert U, et al. Stabilization of transgene expression by incorporation of E3 region genes into an adenoviral factor IX vector and by transient anti-CD4 treatment of the host. Gene Ther 1996;3(6):521 –530. w72x Chen HH, Mack LM, Kelly R, Ontell M, Kochanek S, Clemens PR. Persistence in muscle of an adenoviral vector that lacks all viral genes. Proc Natl Acad Sci USA 1997;94(5):1645 – 1650. w73x Tripathy SK, Black HB, Goldwasser E, Leiden JM. Immune responses to transgene-encoded proteins limit the stability of gene expression after injection of replication-defective adenovirus vectors. Nat Med 1996;2(5):545 –550. w74x Fields PA, Kowalczyk DW, Arruda VR, et al. Role of vector in activation of T cell subsets in immune responses against the secreted transgene product factor IX. Mol Ther 2000;1(3):225 – 235. w75x Suzuki M, Singh R, Moore MA, Song WR, Crystal RG. Similarity of strain- and route-dependent murine responses to an adenovirus vector using the homologous thrombopoietin cDNA as the reporter genes. Hum Gene Ther 1998;9(8):1223 – 1231. w76x Casares S, Inaba K, Brumeanu TD, Steinman RM, Bona CA. Antigen presentation by dendritic cells after immunization with DNA encoding a major histocompatibility complex class IIrestricted viral epitope. J Exp Med 1997;186(9):1481 –1486. w77x Chattergoon MA, Robinson TM, Boyer JD, Weiner DB. Specific immune induction following DNA-based immunization through in vivo transfection and activation of macrophagesy antigen-presenting cells. J Immunol 1998;160(12):5707 –5718. w78x Bouloc A, Walker P, Grivel JC, Vogel JC, Katz SI. Immunization through dermal delivery of protein-encoding DNA: a role for migratory dendritic cells. Eur J Immunol 1999;29(2):446 –454. w79x Porgador A, Irvine KR, Iwasaki A, Barber BH, Restifo NP, Germain RN. Predominant role for directly transfected dendritic cells in antigen presentation to CD8q T cells after gene gun immunization. J Exp Med 1998;188(6):1075 –1082. w80x Ballas ZK, Rasmussen WL, Krieg AM. Induction of NK activity in murine and human cells by CpG motifs in oligodeoxynucleotides and bacterial DNA. J Immunol 1996;157(5):1840 –1845. w81x Klinman DM, Yi AK, Beaucage SL, Conover J, Krieg AM. CpG motifs present in bacteria DNA rapidly induce lymphocytes to secrete interleukin 6, interleukin 12 and interferon gamma. Proc Natl Acad Sci USA 1996;93(7):2879 –2883.
w82x Scheule RK. The role of CpG motifs in immunostimulation and gene therapy. Adv Drug Deliv Rev 2000;44(2–3):119 – 134. w83x Yi AK, Tuetken R, Redford T, Waldschmidt M, Kirsch J, Krieg AM. CpG motifs in bacterial DNA activate leukocytes through the pH-dependent generation of reactive oxygen species. J Immunol 1998;160(10):4755 –4761. w84x Macfarlane DE, Manzel L. Antagonism of immunostimulatory CpG-oligodeoxynucleotides by quinacrine, chloroquine and structurally related compounds. J Immunol 1998;160(3):1122 – 1131. w85x Yi AK, Krieg AM. CpG DNA rescue from anti-IgM-induced WEHI-231 B lymphoma apoptosis via modulation of I kappa B alpha and I kappa B beta and sustained activation of nuclear factor-kappa Byc-Rel. J Immunol 1998;160(3):1240 –1245. w86x Yi AK, Krieg AM. Rapid induction of mitogen-activated protein kinases by immune stimulatory CpG DNA. J Immunol 1998;161(9):4493 –4497. w87x Hacker H, Mischak H, Miethke T, et al. CpG-DNA-specific activation of antigen-presenting cells requires stress kinase activity and is preceded by non-specific endocytosis and endosomal maturation. EMBO J 1998;17(21):6230 –6240. w88x Yamamoto S, Kuramoto E, Shimada S, Tokunaga T. In vitro augmentation of natural killer cell activity and production of interferon-alphaybeta and -gamma with deoxyribonucleic acid fraction from Mycobacterium bovis BCG. Jpn J Cancer Res 1988;79(7):866 –873. w89x Yi AK, Chace JH, Cowdery JS, Krieg AM. IFN-gamma promotes IL-6 and IgM secretion in response to CpG motifs in bacterial DNA and oligodeoxynucleotides. J Immunol 1996;156(2):558 –564. w90x Weiner GJ, Liu HM, Wooldridge JE, Dahle CE, Krieg AM. Immunostimulatory oligodeoxynucleotides containing the CpG motif are effective as immune adjuvants in tumor antigen immunization. Proc Natl Acad Sci USA 1997;94(20):10833 – 10837. w91x Roman M, Martin-Orozco E, Goodman JS, et al. Immunostimulatory DNA sequences function as T helper-1-promoting adjuvants. Nat Med 1997;3(8):849 –854. w92x Sun S, Zhang X, Tough DF, Sprent J. Type I interferonmediated stimulation of T cells by CpG DNA. J Exp Med 1998;188(12):2335 –2342. w93x Chu RS, Targoni OS, Krieg AM, Lehmann PV, Harding CV. CpG oligodeoxynucleotides act as adjuvants that switch on T helper 1 (Th1) immunity. J Exp Med 1997;186(10):1623 – 1631. w94x McDonald D, Stockwin L, Matzow T, Blair Zajdel ME, Blair GE. Coxsackie and adenovirus receptor (CAR)-dependent and major histocompatibility complex (MHC) class I-independent uptake of recombinant adenoviruses into human tumour cells. Gene Ther 1999;6(9):1512 –1519. w95x Fechner H, Haack A, Wang H, et al. Expression of coxsackie adenovirus receptor and alphav-integrin does not correlate with adenovector targeting in vivo indicating anatomical vector barriers. Gene Ther 1999;6(9):1520 –1535. w96x Tallone T, Malin S, Samuelsson A, et al. A mouse model for adenovirus gene delivery. Proc Natl Acad Sci USA 2001;98(14):7910 –7915. w97x Kimura E, Maeda Y, Arima T, et al. Efficient repetitive gene delivery to skeletal muscle using recombinant adenovirus vector containing the coxsackievirus and adenovirus receptor cDNA. Gene Ther 2001;8(1):20 –27. w98x Leissner P, Legrand V, Schlesinger Y, et al. Influence of adenoviral fiber mutations on viral encapsidation, infectivity and in vivo tropism. Gene Ther 2001;8(1):49 –57.
D. Chen et al. / Transplant Immunology 9 (2002) 301–314 w99x Magnusson MK, Hong SS, Boulanger P, Lindholm L. Genetic retargeting of adenovirus: novel strategy employing ‘deknobbing’ of the fiber. J Virol 2001;75(16):7280 –7289. w100x Nettelbeck DM, Miller DW, Jerome V, et al. Targeting of adenovirus to endothelial cells by a bispecific single-chain diabody directed against the adenovirus fiber knob domain and human endoglin (CD105). Mol Ther 2001;3(6):882 –891. w101x Connor RJ, Engler H, Machemer T, et al. Identification of polyamides that enhance adenovirus-mediated gene expression in the urothelium. Gene Ther 2001;8(1):41 –48. w102x Levy RJ, Song C, Tallapragada S, et al. Localized adenovirus gene delivery using antiviral IgG complexation. Gene Ther 2001;8(9):659 –667. w103x Sugai J, Eiden M, Anderson MM, Van Hoeven N, Meiering CD, Overbaugh J. Identification of envelope determinants of feline leukemia virus subgroup B that permit infection and gene transfer to cells expressing human Pit1 or Pit2. J Virol 2001;75(15):6841 –6849. w104x Martin F, Kupsch J, Takeuchi Y, Russell S, Cosset FL, Collins M. Retroviral vector targeting to melanoma cells by singlechain antibody incorporation in envelope. Hum Gene Ther 1998;9(5):737 –746. w105x Engelstadter M, Buchholz CJ, Bobkova M, et al. Targeted gene transfer to lymphocytes using murine leukaemia virus vectors pseudotyped with spleen necrosis virus envelope proteins. Gene Ther 2001;8(15):1202 –1206. w106x Pizzato M, Blair ED, Fling M, et al. Evidence for nonspecific adsorption of targeted retrovirus vector particles to cells. Gene Ther 2001;8(14):1088 –1096. w107x Cosset FL, Morling FJ, Takeuchi Y, Weiss RA, Collins MK, Russell SJ. Retroviral retargeting by envelopes expressing an N-terminal binding domain. J Virol 1995;69(10):6314 –6322. w108x Chadwick MP, Morling FJ, Cosset FL, Russell SJ. Modification of retroviral tropism by display of IGF-I. J Mol Biol 1999;285(2):485 –494. w109x Colin M, Maurice M, Trugnan G, et al. Cell delivery, intracellular trafficking and expression of an integrin-mediated gene transfer vector in tracheal epithelial cells. Gene Ther 2000;7(2):139 –152. w110x Condon C, Watkins SC, Celluzzi CM, Thompson K, Falo LD. DNA-based immunization by in vivo transfection of dendritic cells. Nat Med 1996;2(10):1122 –1128. w111x Ulmer JB, Deck RR, Dewitt CM, Donnhly JI, Liu MA. Generation of MHC class I-restricted cytotoxic T lymphocytes by expression of a viral protein in muscle cells: antigen presentation by non-muscle cells. Immunology 1996;89(1):59 – 67. w112x Doe B, Selby M, Barnett S, Baenziger J, Walker CM. Induction of cytotoxic T lymphocytes by intramuscular immunization with plasmid DNA is facilitated by bone marrow-derived cells. Proc Natl Acad Sci USA 1996;93(16):8578 –8583. w113x Cayeux S, Beck C, Dorken B, Blankenstein T. Coexpression of interleukin-4 and B7.1 in murine tumor cells leads to improved tumor rejection and vaccine effect compared to single gene transfectants and a classical adjuvant. Hum Gene Ther 1996;7(4):525 –529. w114x Chen W, Carbone FR, McCluskey J. Electroporation and commercial liposomes efficiently deliver soluble protein into the MHC class I presentation pathway. Priming in vitro and in vivo for class I-restricted recognition of soluble antigen. J Immunol Methods 1993;160(1):49 –57. w115x van Binnendijk RS, van Baalen CA, Poelen MC, et al. Measles virus transmembrane fusion protein synthesized de novo or presented in immunostimulating complexes is endogenously processed for HLA class I- and class II-restricted cytotoxic T cell recognition. J Exp Med 1992;176(1):119 –128.
313
w116x Kukowska-Latallo JF, Bielinska AU, Johnson J, Spindler R, Tomalia DA, Baker JR. Efficient transfer of genetic material into mammalian cells using Starburst polyamidoamine dendrimers. Proc Natl Acad Sci USA 1996;93(10):4897 –4902. w117x Choi SY, van de Mark K, Faller DV. Identification of a cisacting element in the class I major histocompatibility complex gene promoter responsive to activation by retroviral sequences. J Virol 1997;71(2):965 –970. w118x Suzuki K, Mori A, Ishii KJ, et al. Activation of target-tissue immune-recognition molecules by double-stranded polynucleotides. Proc Natl Acad Sci USA 1999;96(5):2285 –2290. w119x Meijer I, Boot AJ, Mahabir G, Zantema A, van der Eb AJ. Reduced binding activity of transcription factor NF-kappa B accounts for MHC class I repression in adenovirus type 12 E1-transformed cells. Cell Immunol 1992;145(1):56 –65. w120x Burgert HG, Kvist S. An adenovirus type 2 glycoprotein blocks cell surface expression of human histocompatibility class I antigens. Cell 1985;41(3):987 –997. w121x Beier DC, Cox JH, Vining DR, Cresswell P, Engelhard VH. Association of human class I MHC alleles with the adenovirus E3y19K protein. J Immunol 1994;152(8):3862 –3872. w122x Efrat S, Fejer G, Brownlee M, Horwitz MS. Prolonged survival of pancreatic islet allografts mediated by adenovirus immunoregulatory transgenes. Proc Natl Acad Sci USA 1995;92(15):6947 –6951. w123x Lee MG, Abina MA, Haddada H, Perricaudet M. The constitutive expression of the immunomodulatory gp19k protein in E1-, E3-adenoviral vectors strongly reduces the host cytotoxic T cell response against the vector. Gene Ther 1995;2(4):256 – 262. w124x Berger C, Xuereb S, Johnson DC, et al. Expression of herpes simplex virus ICP47 and human cytomegalovirus US11 prevents recognition of transgene products by CD8(q) cytotoxic T lymphocytes. J Virol 2000;74(10):4465 –4473. w125x Yei S, Mittereder N, Wert S, Whitsett JA, Wilmott RW, Trapnell BC. In vivo evaluation of the safety ofadenovirus-mediated transfer of the human cystic fibrosis transmembrane conductance regulator cDNA to the lung. Hum Gene Ther 1994;5(6):731 –744. w126x Adesanya MR, Redman RS, Baum BJ, O’Connell BC. Immediate inflammatory responses to adenovirus-mediated gene transfer in rat salivary glands. Hum Gene Ther 1996;7(9):1085 –1093. w127x Worgall S, Leopold PL, Wolff G, Ferris B, Van Roijen N, Crystal RG. Role of alveolar macrophages in rapid elimination of adenovirus vectors administered to the epithelial surface of the respiratory tract. Hum Gene Ther 1997;8(14):1675 –1684. w128x Worgall S, Wolff G, Falck-Pedersen E, Crystal RG. Innate immune mechanisms dominate elimination of adenoviral vectors following in vivo administration. Hum Gene Ther 1997;8(1):37 –44. w129x Lieber A, He CY, Meuse L, et al. The role of Kupffer cell activation and viral gene expression in early liver toxicity after infusion of recombinant adenovirus vectors. J Virol 1997;71(11):8798 –8807. w130x Medzhitov R, Janeway C. Innate immunity. N Engl J Med 2000;343(5):338 –344. w131x Muruve DA, Barnes MJ, Stillman IE, Liebermann TA. Adenoviral gene therapy leads to rapid induction of multiple chemokines and acute neutrophil-dependent hepatic injury in vivo. Hum Gene Ther 1999;10:965 –976. w132x Siegal FP, Kadowaki N, Shodell M, et al. The nature of the principal type 1 interferon-producing cells in human blood. Science 1999;284(5421):1835 –1837. w133x Siegal FP, Lopez C, Fitzgerald PA, et al. Opportunistic infections in acquired immune deficiency syndrome result from
314
w134x w135x
w136x
w137x
w138x
w139x
w140x
w141x
w142x
w143x
w144x
w145x
w146x
D. Chen et al. / Transplant Immunology 9 (2002) 301–314 synergistic defects of both the natural and adaptive components of cellular immunity. J Clin Invest 1986;78(1):115 –123. Fitzgerald-Bocarsly P. Human natural interferon-alpha producing cells. Pharmacol Ther 1993;60(1):39 –62. Norman J, Denham W, Denham D, et al. Liposome-mediated, nonviral gene transfer induces a systemic inflammatory response which can exacerbate pre-existing inflammation. Gene Ther 2000;7(16):1425 –1430. Schnell MA, Zhang Y, Tazelaar J, et al. Activation of innate immunity in nonhuman primates following intraportal administration of adenoviral vectors. Mol Ther 2001;3(5 Pt1):708 – 722. Otake K, Ennist DL, Harrod K, Trapnell BC. Nonspecific inflammation inhibits adenovirus-mediated pulmonary gene transfer and expression independent of specific acquired immune responses. Hum Gene Ther 1998;9(15):2207 –2222. Lusky M, Christ M, Rittner K, et al. In vitro and in vivo biology of recombinant adenovirus vectors with E1, E1yE2A, or E1yE4 deleted. J Virol 1998;72(3):2022 –2032. Gangappa S, Babu JS, Thomas J, Daheshia M, Rouse BT. Virus-induced immunoinflammatory lesions in the absence of viral antigen recognition. J Immunol 1998;161(8):4289 –4300. Guidotti LG, Borrow P, Brown A, McClary H, Koch R, Chisari FV. Noncytopathic clearance of lymphocytic choriomeningitis virus from the hepatocyte. J Exp Med 1999;189(10):1555 – 1564. Heim A, Zeuke S, Weiss S, Ruschewski W, Grumbach IM. Transient induction of cytokine production in human myocardial fibroblasts by coxsackievirus B3. Circ Res 2000;86(7):753 –759. Amin R, Wilmott R, Schwarz Y, Trapnell B, Stark J. Replication-deficient adenovirus induces expression of interleukin-8 by airway epithelial cells in vitro. Hum Gene Ther 1995;6(2):145 –153. Qin L, Ding Y, Pahud DR, Chang E, Imperiale MJ, Bromberg JS. Promoter attenuation in gene therapy: interferon-gamma and tumor necrosis factor-alpha inhibit transgene expression. Hum Gene Ther 1997;8(17):2019 –2029. Olszewska-Pazdrak B, Casola A, Saito T, et al. Cell-specific expression of RANTES, MCP-1 and MIP-1alpha by lower airway epithelial cells and eosinophils infected with respiratory syncytial virus. J Virol 1998;72(6):4756 –4764. Thomas LH, Friedland JS, Sharland M, Becker S. Respiratory syncytial virus-induced RANTES production from human bronchial epithelial cells is dependent on nuclear factor-kappa B nuclear binding and is inhibited by adenovirus-mediated expression of inhibitor of kappa B alpha. J Immunol 1998;161(2):1007 –1016. Elkon KB, Liu CC, Gall JG, et al. Tumor necrosis factor alpha plays a central role in immune-mediated clearance of adeno-
w147x
w148x
w149x
w150x
w151x
w152x
w153x
w154x
w155x
w156x
w157x
w158x
w159x
viral vectors. Proc Natl Acad Sci USA 1997;94(18):9814 – 9819. Lieber A, He CY, Meuse L, Himeda C, Wilson C, Kay MA. Inhibition of NF-kappaB activation in combination with bcl-2 expression allows for persistence of first-generation adenovirus vectors in the mouse liver. J Virol 1998;72(11):9267 –9277. Soares MP, Muniappan A, Kaczmarek E, et al. Adenovirusmediated expression of a dominant negative mutant of p65y RelA inhibits proinflammatory gene expression in endothelial cells without sensitizing to apoptosis. J Immunol 1998;161(9):4572 –4582. Bohuslav J, Kravchenko VV, Parry GC, et al. Regulation of an essential innate immune response by the p50 subunit of NF-kappaB. J Clin Invest 1998;102(9):1645 –1652. Addison CL, Hitt M, Kunsken D, Graham FL. Comparison of the human versus murine cytomegalovirus immediate early gene promoters for transgene expression by adenoviral vectors. J Gen Virol 1997;78(Pt 7):1653 –1661. Brough DE, Hsu C, Kulesa VA, et al. Activation of transgene expression by early region 4 is responsible for a high level of persistent transgene expression from adenovirus vectors in vivo. J Virol 1997;71(12):9206 –9213. Presti RM, Pollock JL, Dal Canto AJ, O’Guin AK, Virgin 4th HW. Interferon gamma regulates acute and latent murine cytomegalovirus infection and chronic disease of the great vessels. J Exp Med 1998;188(3):577 –588. Lucin P, Jonjic S, Messerle M, Polic B, Hengel H, Koszinowski UH. Late phase inhibition of murine cytomegalovirus replication by synergistic action of interferon-gamma and tumour necrosis factor. J Gen Virol 1994;75(Pt 1):101 –110. Harms JS, Splitter GA. Interferon-gamma inhibits transgene expression driven by SV40 or CMV promoters but augments expression driven by the mammalian MHC I promoter. Hum Gene Ther 1995;6(10):1291 –1297. Gribaudo G, Ravaglia S, Caliendo A, et al. Interferons inhibit onset of murine cytomegalovirus immediate-early gene transcription. Virology 1993;197(1):303 –311. Guidotti LG, Ishikawa T, Hobbs MV, Matzke B, Schreiber R, Chisari FV. Intracellular inactivation of the hepatitis B virus by cytotoxic T lymphocytes. Immunity 1996;4(1):25 –36. Ghazizadeh S, Carroll JM, Taichman LB. Repression of retrovirus-mediated transgene expression by interferons: implications for gene therapy. J Virol 1997;71(12):9163 –9169. Loser P, Jennings GS, Strauss M, Sandig V. Reactivation of the previously silenced cytomegalovirus major immediate-early promoter in the mouse liver: involvement of NFkappaB. J Virol 1998;72(1):180 –190. Hottiger MO, Felzien LK, Nabel GJ. Modulation of cytokineinduced HIV gene expression by competitive binding of transcription factors to the coactivator p300. EMBO J 1998;17(11):3124 –3134.