Available online at www.sciencedirect.com
Transplantation Reviews 23 (2009) 159 – 170 www.elsevier.com/locate/trre
Gene therapy in transplantation Jerome M. Laurencea , Richard D.M. Allena , Geoffrey W. McCaughanb , Grant J. Loganc , Ian E. Alexanderc,d , G. Alex Bishopa,b , Alexandra F. Sharlanda,⁎ a
Collaborative Transplantation Research Group, Bosch Insitute, Royal Prince Alfred Hospital and University of Sydney, 2006, Australia b AW Morrow Liver Immunobiology Laboratory, Centenary Institute, Sydney, 2050, Australia c Gene Therapy Research Unit, Children's Medical Research Institute and The Children's Hospital at Westmead, Westmead, 2145, Australia d Discipline of Paediatrics and Child Health, University of Sydney, 2006, Australia
Abstract Gene therapy is an exciting and novel technology that offers the prospect of improving transplant outcomes beyond those achievable with current clinical protocols. This review explores both the candidate genes and ways in which they have been deployed to overcome both immune and non-immune barriers to transplantation success in experimental models. Finally, the major obstacles to implementing gene therapy in the clinic are considered. © 2009 Elsevier Inc. All rights reserved.
1. Introduction Pharmacologic therapies attempt to correct the disease phenotype, leaving the genotype unaltered. Gene therapy is the delivery of genetic material to a target cell or tissue with the intention of ameliorating disease, seeking to alter the disease-associated genotype. Gene therapy can take various forms. Gene transfer provides the cell with an expressible cloned DNA sequence encoding a functional protein intended to achieve a therapeutic effect. A diseased phenotype might also be improved by reducing the production of certain proteins, a process termed silencing. Gene repair is a process directed at correcting a genetic defect in situ and allowing the corrected gene to be regulated endogenously. Although offering great potential for novel genetic manipulation, gene repair and gene silencing technologies are still in their infancy. This review will therefore focus on gene transfer, a process which requires a safe and efficient means of gene delivery. The characteristics of an ideal gene delivery vector are summarized in Table 1. No single gene delivery system possesses all of these characteristics. Each system will possess a group of properties that dictates its suitability for ⁎ Corresponding author. University of Sydney, NSW 2006, Australia. Tel.: +61 2 9351 2897; fax: +61 2 9036 7083. E-mail address:
[email protected] (A.F. Sharland). 0955-470X/$ – see front matter © 2009 Elsevier Inc. All rights reserved. doi:10.1016/j.trre.2009.04.001
a particular application. Gene transfer technologies, mainly based on viral vectors, are already being used with some success in human therapeutic trials involving cellular autotransplantation [1,2]. However, this review will concentrate on the application of gene delivery technology in solid organ transplantation where, despite extensive experience in small animal studies, no human trials have been conducted. 1.1. Gene therapy in transplantation Transplantation is already a successful treatment of endstage organ failure [3,4]. Acute allograft rejection is effectively prevented by current pharmacotherapies, but half of all grafts are lost by 10 years after transplantation, and long-term treatment is associated with risks of malignancy, vascular disease, and diabetes [5-8]. The major challenges facing patients with organ failure are a shortage of donor organs and, once transplanted, chronic allograft rejection and complications of treatment of rejection. The most advanced efforts toward gene therapy have focused on replacing a deficient gene in the context of a monogenetic disorder [1,2,9,10]. In the context of transplantation, gene therapy offers the prospect of targeted, life-long therapy to prevent rejection. In addition, the modification of organs from other species to render them transplantable into humans and increase access to trans-
160
J.M. Laurence et al. / Transplantation Reviews 23 (2009) 159–170
Table 1 The desirable characteristics of a gene therapy vector Characteristic
Comment
Ease of production Transgene expression profile appropriate to application
High titer preparations should be easy and inexpensive to produce. Target gene expression may need to be sustained (such as when treating phenylketonuria) or may need to be regulated (insulin expression in treatment of diabetes mellitus). An immune response to a vector or transgene may limit the period of effective expression or may promote an immune response to the tissue that expresses it. If the vector delivers the target gene into a specific tissue or cell type, its therapeutic potential may be maximized. The packaging capacity of a vector in terms of size of transgene should not limit therapeutic potential. Ideally dividing cells should continue to express the therapeutic product either by chromosomal integration or formation of an episome that replicates and segregates with cell division. The vector should be able to target cells that are not actively dividing. Host chromosome integration engenders the risk of insertional mutagenesis. This is reduced significantly if integration occurs in specific sites away from critical genes and their regulatory elements.
Mild or no immune response Tissue specificity Vector capacity Continued expression with cell division Ability to target resting as well as dividing cells Site-specific integration
The desirable characteristics of a gene therapy vector [22].
plantation is an important goal. By expressing a therapeutic factor only in the place where it is required, gene therapy may be able to eliminate deleterious effects associated with systemic drug administration. A single dose of a therapeutic gene would be preferable to current pharmacologic therapies, which must be taken regularly and for the life of the graft. Gene therapy also exhibits the potential for the induction of durable, drug-free, immunologic tolerance. Nevertheless, there are many potential problems associated with this modality of treatment, including difficulty regulating transgene expression, safety concerns about malignancy, infection, and inflammation. The application of gene therapy to transplantation only has merit if it can enhance the quantity and quality of life for people with endstage organ failure. 1.2. Modes of gene transfer Genes may be delivered by in vivo gene transfer, where the vector is applied either systemically or locally to the target tissue within the prospective donor or recipient. Alternatively, organs or cells may be collected and modified outside the ultimate host and then returned in a process termed ex vivo gene transfer. Ex vivo transfer limits the exposure of other organs to potentially toxic effects of the vector system, whereas cells modified ex vivo can be selected from those that have failed to undergo modification or expanded in number to allow delivery of a greater inoculum of modified cells. In the realm of solid organ transplantation, whole organs are uniquely accessible for modification ex vivo. However, gene transfer in vivo may be more efficient than ex vivo modification of an organ that is stored cold before transplantation [11]. 1.3. Types of gene delivery vector In general, transfer of DNA into mammalian cells can be readily achieved by a variety of physical (such as pressure or injection), chemical (such as calcium phosphate), or
electrical methods [12,13], collectively known as transfection. Transfection can also be achieved using complexes of nucleic acid and cationic polymers (such as polyamines) known as polyplex systems [14]. Lipid based gene delivery systems include lipid encapsidation of a DNA construct (liposome), cationic lipid and nucleic acid complexes (lipoplexes) [15], and lipopolyplex hybrids of polycationic polymers and lipid [16]. Receptor-mediated uptake of nonviral DNA vectors can be achieved by binding to specific receptors via the conjugation of specific ligands usually to the free amino groups of polycations in the vector and has been shown to increase the efficiency of gene delivery both in vivo and in vitro [17]. The transfer of genetic material using recombinant viruses is known as transduction. Recombinant viral vectors achieve more efficient delivery of nucleic acid than nonviral vectors on a “per molecule” basis [18-23]. Transduction is fundamentally different from infection by a wild-type virus. Transduction is a nonreplicative process leading to the introduction of genetic material into the target cell by the recombinant vector. The only wild-type viral sequences necessarily retained in the recombinant vector genome are those required for genome replication and packaging during vector production and for desirable aspects of the parent viruses' biology. Such sequences are referred to as cis-acting because they regulate the expression of genes on the same nucleic acid strand. Most, or all coding (trans-acting) sequences, are removed. In designing a vector, the therapeutic gene cassette replaces trans-acting elements of the viral genome while maintaining the cis-acting elements (Fig. 1). This generates a vector incapable of replication. The viral coding sequences required for the assembly of viral particles in the packaging cells are provided in trans expressed from heterologous plasmids or incorporated into the producer cell genome. The most commonly used viral vectors in the gene therapy field are derived from adenoviruses (AdVs), gammaretroviruses (RVs), lentiviruses (LVs), and adeno-associated viruses (AAVs). Each system
J.M. Laurence et al. / Transplantation Reviews 23 (2009) 159–170
161
Fig. 1. Converting a wild-type virus into a vector. A, The genome of the virus includes the genes required for replication (blue), viral structural proteins (red), and genes involved in viral pathogenicity (yellow). Flanking these are the viral inverted terminal repeats (ITR), which include the viral origin of replication and packaging signal. The latter sequences are cis-acting as they affect only genes on the same nucleic acid strand. B, The packaging construct, illustrated here as a plasmid, includes the gene required for the production of viral structural proteins (red) and viral replication (blue). These genes are trans-acting as they need not be located on the same nucleic acid strand as the genes that they regulate. C, The recombinant vector contains a transgene cassette with its own transcriptional regulatory elements. This is reproduced and then packaged in the presence of the appropriate viral genes (supplied in trans from the packaging construct) as it is flanked by the necessary cis-acting regulatory sequences. The genes involved in viral pathogenesis are eliminated.
has a unique set of properties that lend themselves to specific applications. For example, RV and LV vectors undergo genomic integration and are therefore favored for applications requiring long-term expression, whereas AdV vectors do not undergo integration and remain as episomes that are rapidly lost in replicating target cell populations with resultant transient expression.
2. Strategies in transplantation gene therapy Efforts thus far at gene therapy in solid organ transplantation have been confined to animal models. In broad terms,
2 strategies have been pursued: modifying the graft and modifying the host or recipient. 2.1. Graft modification Various gene delivery strategies, including both viral and nonviral tools, have been used for both ex vivo and in vivo graft manipulation. Although the graft or the recipient may be the focus of gene delivery, in many cases (particularly where the graft is delivering a factor systemically), gene delivery to the graft exerts its effect most potently through host modification. The chief goal of gene delivery to the graft is the amelioration of insults that are suffered by a transplant
162
J.M. Laurence et al. / Transplantation Reviews 23 (2009) 159–170
as a result of adaptive immune responses, and those that result from innate immune damage, particularly when associated with ischemia and reperfusion injury. 2.1.1. Modulation of co-stimulatory pathways Blockade of co-stimulatory pathways has been extensively explored in many models. The CD28/B7 costimulatory pathway has been targeted using a construct consisting of cytotoxic T-lymphocyte associated antigen 4 (CTLA4) combined with immunoglobulin G. The resulting protein (CTLA4-Ig) is soluble and has a prolonged half-life compared to CTLA4. Using mostly AdV vectors and CTLA4-Ig, prolonged allograft survival has been achieved in rat liver [24,25], rat heart [26-28], rat kidney [29], rat cornea [30], mouse and rat pancreatic islet [31,32], rat lung [33], and rat hind limb models [34]. CTLA4-Ig gene therapy has also been successful in achieving prolonged graft survival in a number of concordant xeno-transplant models including rat-to-mouse islet grafts [35], rat fetal cardiomocytes transplanted into mice [36], and hamster-to-rat liver transplants [37]. In a number of studies, CTLA4-Ig gene therapy has been shown to have synergistic effects with other agents such as immunosuppressive drugs [38], donor leukocyte infusion [39], or concomitant CD40-Ig gene therapy [34,40,41]. The CD40/CD154 co-stimulatory pathway has been a target of gene therapy via the expression of a CD40-Ig fusion protein. Delivered mostly by AdV, this has been successful in prolonging survival of allografts including those of rodent liver [42,43], rodent heart [40,44], hind limb [34], and pancreatic islets [45]. This approach has also been used
successfully in rodent xenograft models of heart and islet transplantation, when combined with immunosuppression (FK779) [46] or concomitant CTLA4-Ig gene therapy [41], respectively. When activation of these co-stimulatory signals has been blocked, the activity of other pathways such as the inducible co-stimulator–B7 related protein-1 pathway has been identified as important in transplant rejection [47]. Although gene therapy to block these additional stimulatory pathways has not so far been successful [48], another pathway that has been targeted with some success is the inhibitory interaction between Programmed Death 1 (PD1) and its ligand Programmed Death 1 Ligand 1 (PD-L1). Heart allografts modified to express a PD-L1-Ig fusion protein have a slight survival advantage over untransduced allografts. This effect was synergistic with low dose immunosuppression [49]. 2.1.2. Manipulation of cytokine expression Certain cytokines or their receptors, particularly interleukin (IL)-10, a virally derived analogue of IL-10 (vIL-10), transforming growth factor β (TGF-β), the p40 subunit of IL-12, or soluble IL-1 receptor, have all been delivered to a graft in the context of transplantation. Interleukin 10 has been the most extensively examined, and there have been relatively smaller numbers of studies focusing on the use of other cytokines for organ modification (Table 2). IL-10 or vIL-10 have produced mostly modest improvements in graft survival in models including nonvascularized cardiac allografts [53,54,63,64], vascularized heterotopic cardiac allografts [52,65-71], liver allografts [72-74], corneal allografts [75,] and pancreatic allografts [76].
Table 2 Studies examining the effects of cytokines for organ modification Gene
Modality of gene therapy
Outcome
Model
Reference
TGF-β
Liposomal transfection and AdV AdV
Mouse vascularized cardiac allograft
[50]
Rat liver allograft
[51]
Rabbit heart allograft Mouse nonvascularized cardiac allograft Mouse vascularized cardiac allograft Human islets transplanted into NOD/SCID mice
[52] [53,54] [55] [56]
Rat in situ I/R model
[57]
Rat vascularized cardiac allograft
[58] [59]
[62]
IL-1R type 2 Ig fusion protein
AdV
IL-13
AdV
P40 subunit of IL-12
RV
Prolonged allograft survival only when combined with CD8+ depletion Reduced TNF-α and IFN-γ production Prolonged allograft survival Prolonged allograft survival Prolonged allograft survival Improved graft function when combined with vascular endothelial growth factor Improved survival and function after ischemia/reperfusion injury Improved survival when combined with immunosuppression Improved survival and function after ischemia/reperfusion injury Prolonged survival
IL-18 binding protein Ig fusion protein IL-17 receptor Ig fusion protein
AdV
Prolonged allograft survival
Rat in situ and syngeneic transplant I/R model Mouse myoblast cell line transplanted into allogeneic strain Rat vascularized cardiac allograft
AdV
Prolonged allograft survival
Rat vascularized cardiac allograft
IL-2 IL-1 receptor antagonist
AdV Plasmid transfection ASO knockdown AdV
AdV or liposomal
TNF-α indicates tumor necrosis factor α; IFN-γ, interferon γ; ASO, antisense oligonucleotide; I/R, ischemia and reperfusion.
[60] [61]
J.M. Laurence et al. / Transplantation Reviews 23 (2009) 159–170
163
Table 3 Studies examining the effects of expression of leukocyte adhesion molecules and chemokines Gene
Modality of gene therapy
Outcome
Model
Reference
vMIP-II and MC148
Plasmid transfection
Prolonged allograft survival
[93]
ICAM-1
Hyperbaric injection of ASO leading to ICAM-1 knockdown ASO injection leading to ICAM-1 knockdown Hyperbaric injection of ASO leading to ICAM-1 knockdown LV expressing chemokine antagonist RANTES 9-68 AdV expressing NH(2)-terminal deletion mutants of MCP-1 and RANTES
Reduced ischemia and reperfusion injury Prolonged survival
Mouse vascularized and nonvascularized cardiac allograft Rat heterotopic cardiac allograft Mouse islet allograft
[90]
RANTES MCP-1 and RANTES
[89]
Prolonged survival and reduced chronic rejection Prolonged survival
Rat heterotopic cardiac allograft
[94]
Rat heterotopic cardiac allograft
[91]
Prolonged survival
Rat heterotopic cardiac allograft
[92]
ASO indicates anti-sense oligonucleotide; MCP-1, monocyte chemoattractant protein 1.
In the context of lung allografts, improved function after transplantation was observed using IL-10–mediated gene therapy [77-81]. Although IL-10 gene transfer to pancreatic islet transplants produced prolonged survival when returned to NOD syngeneic hosts [82,83], no protection was afforded when transplanted to allogeneic recipients [84]. The modest allograft survival prolongation achieved by IL-10 gene therapy when used alone was greatly augmented by the addition of concomitant IL-4 gene therapy [85]. A variety of mechanisms may contribute to the graft protection afforded by the local expression of IL-10 or its viral analogue. Graft IL-10 or vIL-10 expression reduces infiltration by both CD4+ and CD8+ T cells [54,65,67], while decreasing the expression of proinflammatory cytokines, such as interferon γ and tumor necrosis factor [67,77]. Interleukin 10 increased the apoptosis of alloreactive CD8+ T cells infiltrating the graft possibly through activationinduced death via the Fas/Fas-L and Bax pathways [86,87]. The protection of aortic allografts from chronic rejection afforded by IL-10 gene therapy was shown to be mediated by reduced neointimal proliferation mediated through IL-10– dependent up-regulation of hemoxygenase 1 (HO-1) [70]. Interleukin 10 is known to induce Treg [88], and it is tempting to speculate that the effects of gene therapy using IL-10 may involve this mechanism. However, there is no direct evidence to support such a contention. 2.1.3. Leukocyte recruitment and adhesion A number of studies have focused on modulating rejection by altering leukocyte tethering to the inflamed endothelium of the transplanted tissue. In particular, attempts have been made to disrupt the interaction between intercellular adhesion molecule 1 (ICAM-1) and its integrin receptor, lymphocyte function-associated antigen 1, using antisense oligonucleotide knockdown of ICAM-1 expression. This has been shown to reduce ischemia and reperfusion injury associated with heterotopic rat heart allografts [89], prolong survival of mouse islet allografts [90], and reduce chronic rejection of heterotopic rat heart transplants. Attempts have been made to disrupt the
ability of chemokines to attract and activate leukocytes to the site of inflammation within the graft by using antagonists of chemokine-mediated attraction. Specifically, antagonists of the CC-chemokine regulated on activation, normal T cell expressed and secreted (RANTES) [91,92], and viral chemokine antagonists viral macrophage inflammatory protein II (vMIP-II) and MC148 [93], have been delivered using gene therapy techniques to prolong survival of heterotopic heart allografts (Table 3). 2.1.4. Immunomodulatory and cytoprotective enzymes Attempts have been made to use gene therapy to exploit the immunomodulatory and cytoprotective capacity of enzymes including indoleamine dioxygenase (IDO), HO-1, nitric oxide synthetase, superoxide dismutase, and catalase. Indoleamine dioxygenase expression has been shown to slightly prolong the survival of corneal allografts [95] and to attenuate acute injury [96] and chronic fibrosis [97] after lung allo-transplantation. However, high-level expression of IDO in liver using AAV has no effect on liver transplant rejection [98]. Nitric oxide synthetase gene transfer to arterial allografts protects them from rejection [99-101]. Hemoxygenase 1, when administered systemically via an AdV vector, produces long-term survival of rat heart allografts [102]. Adenovirus HO-1 gene transfer protects against I/R injury in steatotic rat livers by preventing apoptosis of injured cells and prolongs rat liver allograft survival modestly [103-107]. Hemoxygenase 1 gene transfer using an AdV vector protects the function and viability of human islets in culture [108] and ameliorates chronic rejection of rat aortic allograft [109]. Superoxide dismutase delivered via an AdV vector attenuates I/R injury and, in rat liver transplantation models, improves survival of recipients of normal and fatty livers [110,111]. Adenovirus transfer of the catalase gene reduced susceptibility to oxidative stress in vitro [112]. After delivery by polylipid nanoparticles, the genes for both superoxide dismutase and catalase were shown to ameliorate the effects of warm in situ ischemic injury in the mouse liver [113].
164
J.M. Laurence et al. / Transplantation Reviews 23 (2009) 159–170
2.1.5. Apoptosis pathways A number of studies have examined the delivery of genes to induce apoptosis of infiltrating alloimmune cells. The expression of Fas-L has produced results that are disparate and difficult to reconcile. Modest prolongation of rat kidney allograft survival was achieved after graft transduction by an AdV expressing Fas-L [114]. Syngeneic myoblasts expressing Fas-L co-transplanted with islet allografts were reported to prolong islet allograft survival in a site-specific manner [115]. However, when pancreatic islets themselves expressed Fas-L (either after AdV transduction or in a transgenic model), those cells were rapidly destroyed by a lymphocyte-independent immune response dominated by IL8 expression and the recruitment of granulocytes [116,117]. This was not a phenomenon affecting pancreatic islets exclusively, as heart grafts expressing Fas-L, even when transplanted into syngeneic recipients, were subject to a rapid and destructive inflammatory response [118]. These apparently contradictory results may be partly explained by differing levels of Fas expression on the parenchymal cells of different organs (parenchymal cells expressing Fas being susceptible to fratricide or suicide upon Fas-L co-expression) and partly by the narrow therapeutic window for this gene [114,119]. These results may also reflect engagement of pattern-recognition receptors in some, but not all, cell types as an unintended consequence of viral transduction. Fas–Fas-L interactions have also been targeted indirectly by gene therapy with IL-10, which promotes apoptosis of alloreactive T cells by increasing their expression of Fas [69,87]. Bcl-2, Bcl-xl, and A20 are antiapoptotic genes that may protect cells targeted by an alloimmune response from nuclear factor κB–mediated immune activation and apoptosis [120,121]. There is evidence to suggest that the combination of A1 with A20 may have a synergistic protective effect [122]. AdV transduction of the liver with Bcl-2 improved its resistance to damage with prolonged cold storage and ex vivo rewarming [123], and transfer of Bcl-xl to the corneal endothelium by a lentiviral vector has been shown to prolong allograft survival [124]. Gene therapy with A20 aids the resistance of pancreatic islets to the insult of procurement and transplantation and results in cure of diabetes in syngeneic diabetic mice with a smaller inoculum of islet cells than usually required [125]. 2.2. Host modification Host modification strategies aim to render a recipient more amenable to accepting an allograft. Many of these techniques are based on exposing the recipient to donor major histocompatibility complex (MHC) encoded by a vector aiming to mimic the effects of donor-leukocyte infusion. Conversely, some groups have sought to enhance the tolerance-inducing capacity of donor cells by genetic modification of donor antigen-presenting cell (APC) (particularly dendritic cell [DC]). This strategy seeks to modify the host immune response without genetic modification of the graft.
2.2.1. Donor MHC transfer The goal of donor MHC transfer is to modify autologous cells to encode donor-type MHC proteins. This may allow the realization of the potential of tolerance to those proteins without the risk of graft-versus-host disease that is associated with tolerance induced by allogeneic bone marrow transplantation [126]. Retroviral transduction of autologous bone marrow to express a class I MHC antigen from an allogenic strain induced specific tolerance to skin grafts derived from animals of that strain by T-cell anergy rather than by clonal deletion [127-129]. Similarly, AdV transfer of class I MHC antigen resulted in long-term survival of heart allografts from the same strain when combined with transient CD4+ T-cell depletion [130]. Introduction of allogeneic MHC class I genes into the thymus either by direct vector injection [131] or by injection of transfected autologous cells [132] produced donor-specific tolerance to subsequent allografts. Transduction of a heart allograft recipient's native liver with an AdV vector expressing soluble allogeneic MHC class I antigen modestly prolonged the survival of subsequent heart allografts sharing the MHC class I antigen encoded by the AdV vector [133]. Delivery of a gene expressing allogeneic MHC class II antigens by retroviral vector into autologous bone marrow cells prolonged survival of allografts sharing the same antigen as the transduced bone marrow but differing at other MHC loci [134,135]. One of the major limiting factors with MHC gene therapy is the maintenance of sufficient autologous cells expressing the allogeneic MHC to achieve durable tolerance. This has been addressed by the addition of a selection marker to the vector expressing allogeneic MHC, enabling selective survival of transduced stem cells and ensuring persistence of allograft tolerance [136]. 2.2.2. Transfer of transduced donor antigen-presenting cells APC can act to augment or down-regulate an immune response depending on the available second signals and the prevailing state of immune activation [137]. The process of solid organ transplantation is intrinsically immunogenic, being associated with ischemia-reperfusion injury and other insults to the graft and recipient. This tends to skew the profile of DC away from a tolerogenic program in a number of ways, including an increase in the expression of costimulatory molecules interacting with the T cell [138]. Viral transduction per se (particularly when AdV vectors are used) can also render APC more immunogenic. Modification of the APC using gene therapy aims to turn the APC toward a tolerogenic program, counteracting the immune activating effects not only of transplantation but also of viral transduction on these cells. A number of studies have modified DC and demonstrated a capacity for reduced allostimulation in vitro from DC expressing a range of transgenes including CTLA-4 [139], vIL-10 [140], IDO [141], and Fas-L [142]. The expression of TGF-β was able to counteract the immunostimulatory effect of AdV transduction on DC [143]. Using donor-derived DC modified
J.M. Laurence et al. / Transplantation Reviews 23 (2009) 159–170
to express IL-10 [144], Fas-L [145], or CTLA-4 [146], the survival of small bowel, heart allografts, and islet allografts, respectively, was modestly prolonged. Conversely, the expression of IL-4 by allogeneic DC augmented immune responses in vitro and accelerated allograft rejection [147]. 2.2.3. Transfer of transduced donor lymphocytes T lymphocyte and natural killer cell subpopulations modulate immune homeostasis and have also been shown to contribute to tolerance induction in transplantation [148]. Markers, such as CTLA4, GITR, and FoxP3, have enabled the identification of T-regulatory cells, and gene therapy has been applied to their generation for tolerance induction in preclinical transplantation studies. Chai et al [149] used retroviral vectors encoding FoxP3, a transcription repressor protein important for regulatory cell function, to transduce T lymphocytes transgenic for the male specific H-Y antigen. Adoptive transfer of the resultant population to female mice significantly prolonged the survival of male skin grafts. In an alternative approach, Tsang et al [150] transduced regulatory T lymphocytes with a retrovirus encoding a TCR specific for an allogeneic peptide presented by MHC class II molecules of the recipient APC. This conferred the regulatory cells with specificity to transplanted cardiac grafts which, in combination with rapamycin and anti-CD8 antibody treatment, significantly enhanced the survival of fully mismatched cardiac grafts. Although highly artificial in their design, these studies demonstrate proof-of-principle for exploitation of gene therapy and regulatory T cells to improve transplantation outcomes in the clinic.
3. Potential clinical application of gene therapy in transplantation Over more than a decade, experimental animal studies have clearly demonstrated success in overcoming both immune and non-immune barriers to organ transplantation using a wide variety of candidate genes, delivered by a range of vectors, using a number of different strategies. However, there are many obstacles to be overcome before these advances can be translated into a treatment that would improve outcomes in human clinical transplantation. Gene therapy has found most ready application in human diseases where the outcomes are currently very poor and conventional treatments either highly toxic or relatively ineffective, or both. Examples included advanced malignancy or severe genetic disorders [151]. However, early human gene therapy trials have suffered a number of well-publicized adverse events. During the trial of an AdV vector to treat a partial ornithine transcarbamylase deficiency, an 18-year-old study participant died of a severe systemic inflammatory response to components of the vector. The severity of this response had not been anticipated based on preceding animal studies or even previous experience in the same study with lower
165
doses of the vector [152]. Another major setback in human gene therapy clinical trials was the development of malignancy in an infant successfully treated for X-linked severe combined immunodeficiency using retroviral gene therapy [153] due to an insertional mutagenesis event. This complication was believed before the trial to be a remote possibility, but malignancy was eventually reported in 4 of 9 patients treated in this trial [154] and in 1 of 10 in a second similar trial [155]. It is therefore likely that only once gene therapy has been successfully established as a safe and effective treatment in another area of medicine that it can be advanced as an alternative or adjuvant to current transplantation protocols. The clinical scenario in which gene therapy is likely to be most appropriate is that of deceased organ donation. Here, it may be feasible to deliver a recombinant vector to a whole organ before implantation either by ex vivo manipulation or by delivery in vivo before organ procurement from the donor. Nevertheless, there are a number of issues surrounding the clinical applicability of these techniques. Given that the different organs have varying tropisms for different vectors and promoter systems [22], it is unlikely that a single vector would be ideal for all the commonly transplanted tissues. Ex vivo gene delivery is a way of avoiding exposure of other organs to a vector or segregating organs such that the most ideal vector system can be used for each tissue. However, perfusion of the cold preserved organ is likely to be less effective than in vivo gene delivery [11] and thus not the preferred technique from the point of view of transduction efficiency. Another issue is the relatively slow kinetics of gene expression mediated by recombinant vectors. For example, although AAV is an otherwise promising vector, maximal expression is achieved only some weeks after vector administration in animal models [22]. The average length of stay in an intensive care unit for a deceased donor is around 3 days [156], and the time from the diagnosis of brain death to organ procurement is even less. Thus, the kinetics of AAV vectors are incompatible with this clinical scenario. However, variant AAV vectors are now available that may help circumvent the requirement for complementary DNA strand synthesis and provide not only more rapid onset of transgene expression [157] but also more efficient transduction [158]. Such self-complementary vectors may enhance the prospects for the clinical applicability of AAV in the context of transplantation. Although AdV vectors achieve more rapid transgene expression than AAV vectors [22], AdV administration causes activation of host parenchymal and myeloid cells independent of AdV gene transcription. The myeloid cells in particular release chemokines, both locally and systemically, in response to AdV transduction [159,160]. This response is associated with cellular inflammation and even fatal systemic inflammatory responses [58]. Not only does this feature of AdV pose safety risks, but it limits the long-term expression of the AdV-encoded genes.
166
J.M. Laurence et al. / Transplantation Reviews 23 (2009) 159–170
4. Conclusions Any novel treatment must offer real advantages with low risk of adverse events. As described above, the possibility of unanticipated complications has been realized with previous attempts to use gene therapy to treat genetic disease. Although many animal studies have been performed using gene therapy in transplantation, most have shown only a modest effect. In some studies, gene therapy has been augmented by the addition of subtherapeutic immunosuppressive drug treatment [38,46,49,58]. However, this approach may also increase the risk of complications (such as malignancy or infection) associated with the use of viral vectors. The latter possibility may hamper the prospects for a graded introduction of gene therapy technologies combined with current treatments. Therefore, the greatest barrier to advancing gene therapy into the clinic is the very success of the current practice of transplantation. This work was supported by the National Health and Medical Research Council of Australia and the Microsearch Foundation of Australia. JML was supported by a scholarship from the Northcote Foundation. No authors have any conflict of interest to declare.
References [1] Cavazzana-Calvo M, Hacein-Bey S, de Saint Basile G, et al. Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease. Science 2000;288:669-72. [2] Aiuti A, Slavin S, Aker M, et al. Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning. Science 2002;296:2410-3. [3] Cecka JM. The OPTN/UNOS Renal Transplant Registry. Clin Transpl 2005:1-16. [4] Futagawa Y, Terasaki PI. An analysis of the OPTN/UNOS Liver Transplant Registry. Clin Transpl 2004:315-29. [5] Cecka J. The UNOS scientific renal transplant registry: ten years of kidney transplants. In: Cecka J, Terasaki P, editors. Clinical Transplants. Los Angeles: UCLA tissue Typing Laboratory; 1997. p. 1-15. [6] Beveridge T, Calne RY. Cyclosporine (Sandimmun) in cadaveric renal transplantation. Ten-year follow-up of a multicenter trial. European Multicentre Trial Group. Transplantation 1995;59: 1568-70. [7] Nagano H, Tilney NL. Chronic allograft failure: the clinical problem. Am J Med Sci 1997;313:305-9. [8] Shah T, Kasravi A, Huang E, et al. Risk factors for development of new-onset diabetes mellitus after kidney transplantation. Transplantation 2006;82:1673-6. [9] Ott MG, Schmidt M, Schwarzwaelder K, et al. Correction of X-linked chronic granulomatous disease by gene therapy, augmented by insertional activation of MDS1-EVI1, PRDM16 or SETBP1. Nat Med 2006;12:401-9. [10] Schwarzwaelder K, Howe SJ, Schmidt M, et al. Gammaretrovirusmediated correction of SCID-X1 is associated with skewed vector integration site distribution in vivo. J Clin Invest 2007;117:2241-9. [11] Takahashi Y, Geller DA, Gambotto A, Watkins SC, Fung JJ, Murase N. Adenovirus-mediated gene therapy to liver grafts: successful gene transfer by donor pretreatment. Surgery 2000;128:345-52.
[12] Mann MJ, Gibbons GH, Hutchinson H, et al. Pressure-mediated oligonucleotide transfection of rat and human cardiovascular tissues. Proc Natl Acad Sci U S A 1999;96:6411-6. [13] Aihara H, Miyazaki J. Gene transfer into muscle by electroporation in vivo. Nat Biotechnol 1998;16:867-70. [14] Goldman CK, Soroceanu L, Smith N, et al. In vitro and in vivo gene delivery mediated by a synthetic polycationic amino polymer. Nat Biotechnol 1997;15:462-6. [15] Felgner PL, Barenholz Y, Behr JP, et al. Nomenclature for synthetic gene delivery systems. Hum Gene Ther 1997;8:511-2. [16] Yamazaki Y, Nango M, Matsuura M, Hasegawa Y, Hasegawa M, Oku N. Polycation liposomes, a novel nonviral gene transfer system, constructed from cetylated polyethylenimine. Gene Ther 2000;7:1148-55. [17] Kren BT, Chowdhury NR, Chowdhury JR, Steer CJ. Gene therapy as an alternative to liver transplantation. Liver Transpl 2002;8:1089-108. [18] Gojo S, Yamamoto S, Patience C, LeGuern C, Cooper DK. Gene therapy—its potential in surgery. Ann R Coll Surg Engl 2002;84: 297-301. [19] Tan PH, Chan CL, Chan C, George AJ. The evolving role of genebased treatment in surgery. Br J Surg 2005;92:1466-80. [20] Nishikawa M, Huang L. Nonviral vectors in the new millennium: delivery barriers in gene transfer. Hum Gene Ther 2001;12:861-70. [21] Somia N, Verma IM. Gene therapy: trials and tribulations. Nat Rev Genet 2000;1:91-9. [22] Verma IM, Weitzman MD. Gene therapy: twenty-first century medicine. Annu Rev Biochem 2005;74:711-38. [23] Boeckle S, Wagner E. Optimizing targeted gene therapy: chemical modification of viral vectors and synthesis of artificial virus vector systems. AAPS J 2006:E731-42. [24] Olthoff KM, Judge TA, Gelman AE, et al. Adenovirus-mediated gene transfer into cold-preserved liver allografts: survival pattern and unresponsiveness following transduction with CTLA4Ig. Nat Med 1998;4:194-200. [25] Yanagida N, Nomura M, Yamashita K, et al. Tolerance induction by a single donor pretreatment with the adenovirus vector encoding CTLA4Ig gene in rat orthotopic liver transplantation. Transplant Proc 2001;33:573-4. [26] Guillot C, Mathieu P, Coathalem H, et al. Tolerance to cardiac allografts via local and systemic mechanisms after adenovirusmediated CTLA4Ig expression. J Immunol 2000;164:5258-68. [27] Kita Y, Li XK, Ohba M, et al. Prolonged cardiac allograft survival in rats systemically injected adenoviral vectors containing CTLA4Iggene. Transplantation 1999;68:758-66. [28] Yang Z, Rostami S, Koeberlein B, Barker CF, Naji A. Cardiac allograft tolerance induced by intra-arterial infusion of recombinant adenoviral CTLA4Ig. Transplantation 1999;67:1517-23. [29] 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;11:747-52. [30] Comer RM, King WJ, Ardjomand N, Theoharis S, George AJ, Larkin DF. Effect of administration of CTLA4-Ig as protein or cDNA on corneal allograft survival. Invest Ophthalmol Vis Sci 2002;43: 1095-103. [31] Laumonier T, Potiron N, Boeffard F, et al. CTLA4Ig adenoviral gene transfer induces long-term islet rat allograft survival, without tolerance, after systemic but not local intragraft expression. Hum Gene Ther 2003;14:561-75. [32] 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;12:415-26. [33] 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;71:1867-71. [34] Kanaya K, Tsuchida Y, Inobe M, et al. Combined gene therapy with adenovirus vectors containing CTLA4Ig and CD40Ig prolongs
J.M. Laurence et al. / Transplantation Reviews 23 (2009) 159–170
[35]
[36]
[37]
[38]
[39]
[40]
[41]
[42]
[43]
[44]
[45]
[46]
[47]
[48]
[49]
[50]
[51]
[52]
survival of composite tissue allografts in rat model. Transplantation 2003;75:275-81. Feng S, Quickel RR, Hollister-Lock J, et al. Prolonged xenograft survival of islets infected with small doses of adenovirus expressing CTLA4Ig. Transplantation 1999;67:1607-13. Li TS, Hamano K, Kajiwara K, Nishida M, Zempo N, Esato K. Prolonged survival of xenograft fetal cardiomyocytes by adenovirusmediated CTLA4-Ig expression. Transplantation 2001;72:1983-5. Guo L, Li X, Enosawa S, et al. Prolongation of liver xenograft survival by adenovirus vector-mediated CTLA-4Ig gene transfer. Transpl Immunol 2003;11:155-62. Yang Z, Wu X, Tsui TY, Hou Y, Luk JM, Fan ST. Long-term liver allograft survival induced by combined treatment with rAAVhCTLA4Ig gene transfer and low-dose FK506. Transplantation 2003;75:303-8. Jin Y, Zhang Q, Hao J, Gao X, Guo Y, Xie S. Simultaneous administration of a low-dose mixture of donor bone marrow cells and splenocytes plus adenovirus containing the CTLA4Ig gene result in stable mixed chimerism and long-term survival of cardiac allograft in rats. Immunology 2003;110:275-86. Yamashita K, Masunaga T, Yanagida N, et al. Long-term acceptance of rat cardiac allografts on the basis of adenovirus mediated CD40Ig plus CTLA4Ig gene therapies. Transplantation 2003;76:1089-96. Jin YZ, Xie SS. Bicistronic adenovirus-mediated gene transfer of CTLA4Ig gene and CD40Ig gene result in indefinite survival of islet xenograft. Transplant Proc 2003;35:3165-6. Chang GJ, Liu T, Feng S, et al. Targeted gene therapy with CD40Ig to induce long-term acceptance of liver allografts. Surgery 2002; 132:149-56. Nomura M, Yamashita K, Murakami M, et al. Induction of donorspecific tolerance by adenovirus-mediated CD40Ig gene therapy in rat liver transplantation. Transplantation 2002;73:1403-10. Guillot C, Guillonneau C, Mathieu P, et al. Prolonged blockade of CD40-CD40 ligand interactions by gene transfer of CD40Ig results in long-term heart allograft survival and donor-specific hyporesponsiveness, but does not prevent chronic rejection. J Immunol 2002; 168:1600-9. Rehman KK, Bertera S, Trucco M, Gambotto A, Robbins PD. Immunomodulation by adenoviral-mediated SCD40-Ig gene therapy for mouse allogeneic islet transplantation. Transplantation 2007; 84:301-7. Hua N, Yamashita K, Hashimoto T, et al. Gene therapy-mediated CD40L and CD28 co-stimulatory signaling blockade plus transient anti-xenograft antibody suppression induces long-term acceptance of cardiac xenografts. Transplantation 2004;78:1463-70. Ozkaynak E, Gao W, Shemmeri N, et al. Importance of ICOS– B7RP-1 costimulation in acute and chronic allograft rejection. Nat Immunol 2001;2:591-6. Fabian D, Gong N, Vogt K, Volk HD, Pleyer U, Ritter T. The influence of inducible costimulator fusion protein (ICOSIg) gene transfer on corneal allograft survival. Graefes Arch Clin Exp Ophthalmol 2007;245:1515-21. Dudler J, Li J, Pagnotta M, Pascual M, von Segesser LK, Vassalli G. Gene transfer of programmed death ligand-1.Ig prolongs cardiac allograft survival. Transplantation 2006;82:1733-7. Chan SY, Goodman RE, Szmuszkovicz JR, Roessler B, Eichwald EJ, Bishop DK. DNA-liposome versus adenoviral mediated gene transfer of transforming growth factor beta1 in vascularized cardiac allografts: differential sensitivity of CD4+ and CD8+ T cells to transforming growth factor beta1. Transplantation 2000;70:1292-301. Drazan KE, Olthoff KM, Wu L, Shen XD, Gelman A, Shaked A. Adenovirus-mediated gene transfer in the transplant setting: early events after orthotopic transplantation of liver allografts expressing TGF-beta1. Transplantation 1996;62:1080-4. Brauner R, Nonoyama M, Laks H, et al. Intracoronary adenovirusmediated transfer of immunosuppressive cytokine genes prolongs allograft survival. J Thorac Cardiovasc Surg 1997;114:923-33.
167
[53] 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; 59:809-16. [54] Qin L, Ding Y, Bromberg JS. Gene transfer of transforming growth factor-beta 1 prolongs murine cardiac allograft survival by inhibiting cell-mediated immunity. Hum Gene Ther 1996;7:1981-8. [55] Qu X, Kirken RA, Tian L, Wang M, Bennett CF, Stepkowski SM. Selective inhibition of IL-2 gene expression by IL-2 antisense oligonucleotides blocks heart allograft rejection. Transplantation 2001;72:915-23. [56] Narang AS, Sabek O, Gaber AO, Mahato RI. Co-expression of vascular endothelial growth factor and interleukin-1 receptor antagonist improves human islet survival and function. Pharm Res 2006;23:1970-82. [57] Harada H, Wakabayashi G, Takayanagi A, et al. Transfer of the interleukin-1 receptor antagonist gene into rat liver abrogates hepatic ischemia-reperfusion injury. Transplantation 2002;74:1434-41. [58] Simeoni E, Dudler J, Fleury S, et al. Gene transfer of a soluble IL-1 type 2 receptor-Ig fusion protein improves cardiac allograft survival in rats. Eur J Cardiothorac Surg 2007;31:222-8. [59] Ke B, Shen XD, Lassman CR, et al. Interleukin-13 gene transfer protects rat livers from antigen-independent injury induced by ischemia and reperfusion. Transplantation 2003;75:1118-23. [60] 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 U S A 1996;93:9085-9. [61] Dudler J, Simeoni E, Fleury S, et al. Gene transfer of interleukin-18– binding protein attenuates cardiac allograft rejection. Transpl Int 2007;20:460-6. [62] Li J, Simeoni E, Fleury S, et al. Gene transfer of soluble interleukin17 receptor prolongs cardiac allograft survival in a rat model. Eur J Cardiothorac Surg 2006;29:779-83. [63] Qin L, Ding Y, Pahud DR, Robson ND, Shaked A, Bromberg JS. Adenovirus-mediated gene transfer of viral interleukin-10 inhibits the immune response to both alloantigen and adenoviral antigen. Hum Gene Ther 1997;8:1365-74. [64] Fu S, Chen D, Mao X, Zhang N, Ding Y, Bromberg JS. Feline immunodeficiency virus–mediated viral interleukin-10 gene transfer prolongs non-vascularized cardiac allograft survival. Am J Transplant 2003;3:552-61. [65] DeBruyne LA, Li K, Chan SY, Qin L, Bishop DK, Bromberg JS. Lipid-mediated gene transfer of viral IL-10 prolongs vascularized cardiac allograft survival by inhibiting donor-specific cellular and humoral immune responses. Gene Ther 1998;5:1079-87. [66] Wang CK, Zuo XJ, Carpenter D, et al. Prolongation of cardiac allograft survival with intracoronary viral interleukin-10 gene transfer. Transplant Proc 1999;31:951-2. [67] David A, Chetritt J, Guillot C, et al. Interleukin-10 produced by recombinant adenovirus prolongs survival of cardiac allografts in rats. Gene Ther 2000;7:505-10. [68] Hong YS, Laks H, Cui G, Chong T, Sen L. Localized immunosuppression in the cardiac allograft induced by a new liposome-mediated IL-10 gene therapy. J Heart Lung Transplant 2002;21:1188-200. [69] Oshima K, Sen L, Cui G, et al. Localized interleukin-10 gene transfer induces apoptosis of alloreactive T cells via FAS/FASL pathway, improves function, and prolongs survival of cardiac allograft. Transplantation 2002;73:1019-26. [70] Chen S, Kapturczak MH, Wasserfall C, et al. Interleukin 10 attenuates neointimal proliferation and inflammation in aortic allografts by a heme oxygenase–dependent pathway. Proc Natl Acad Sci U S A 2005;102:7251-6. [71] Zhao J, Pettigrew GJ, Bolton EM, et al. Lentivirus-mediated gene transfer of viral interleukin-10 delays but does not prevent cardiac allograft rejection. Gene Ther 2005;12:1509-16.
168
J.M. Laurence et al. / Transplantation Reviews 23 (2009) 159–170
[72] 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;6:816-22. [73] Tashiro H, Shinozaki K, Yahata H, et al. Prolongation of liver allograft survival after interleukin-10 gene transduction 24–48 hours before donation. Transplantation 2000;70:336-9. [74] Hong IC, Mullen PM, Precht AF, et al. Non-viral human IL-10 gene expression reduces acute rejection in heterotopic auxiliary liver transplantation in rats. Microsurgery 2003;23:432-6. [75] Klebe S, Sykes PJ, Coster DJ, Krishnan R, Williams KA. Prolongation of sheep corneal allograft survival by ex vivo transfer of the gene encoding interleukin-10. Transplantation 2001;71:1214-20. [76] Ozden H, Kabay B, Guven G, Acikalin MF, Erbis H, Alatas O. Interleukin-10 gene transfection of donor pancreas grafts protects against rejection after heterotopic pancreas transplantation in a rat model. Eur Surg Res 2005;37:220-7. [77] Fischer S, Liu M, MacLean AA, et al. In vivo transtracheal adenovirus-mediated transfer of human interleukin-10 gene to donor lungs ameliorates ischemia-reperfusion injury and improves early posttransplant graft function in the rat. Hum Gene Ther 2001; 12:1513-26. [78] Itano H, Mora BN, Zhang W, et al. Lipid-mediated ex vivo gene transfer of viral interleukin 10 in rat lung allotransplantation. J Thorac Cardiovasc Surg 2001;122:29-38. [79] de Perrot M, Fischer S, Liu M, et al. Impact of human interleukin-10 on vector-induced inflammation and early graft function in rat lung transplantation. Am J Respir Cell Mol Biol 2003;28:616-25. [80] Fischer S, De Perrot M, Liu M, et al. Interleukin 10 gene transfection of donor lungs ameliorates posttransplant cell death by a switch from cellular necrosis to apoptosis. J Thorac Cardiovasc Surg 2003; 126:1174-80. [81] Shoji F, Yonemitsu Y, Okano S, et al. Airway-directed gene transfer of interleukin-10 using recombinant Sendai virus effectively prevents post-transplant fibrous airway obliteration in mice. Gene Ther 2003; 10:213-8. [82] Zhang YC, Pileggi A, Agarwal A, et al. Adeno-associated virusmediated IL-10 gene therapy inhibits diabetes recurrence in syngeneic islet cell transplantation of NOD mice. Diabetes 2003; 52:708-16. [83] Carter JD, Ellett JD, Chen M, et al. Viral IL-10–mediated immune regulation in pancreatic islet transplantation. Mol Ther 2005; 12:360-8. [84] Zhang YC, Pileggi A, Molano RD, et al. Systemic overexpression of interleukin-10 fails to protect allogeneic islet transplants in nonobese diabetic mice. Transplantation 2005;80:530-3. [85] Furukawa H, Oshima K, Tung T, Cui G, Laks H, Sen L. Liposomemediated combinatorial cytokine gene therapy induces localized synergistic immunosuppression and promotes long-term survival of cardiac allografts. J Immunol 2005;174:6983-92. [86] Oshima K, Cui G, Tung T, Okotie O, Laks H, Sen L. Exogenous IL10 overexpression reduces perforin production by activated allogenic CD8+ cells and prolongs cardiac allograft survival. Am J Physiol Heart Circ Physiol 2007;292:H277-84. [87] Tung TC, Oshima K, Cui G, Laks H, Sen L. Dual upregulation of Fas and Bax promotes alloreactive T cell apoptosis in IL-10 gene targeting of cardiac allografts. Am J Physiol Heart Circ Physiol 2003;285: H964-73. [88] Groux H, O'Garra A, Bigler M, et al. A CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis. Nature 1997; 389:737-42. [89] Feeley BT, Poston RS, Park AK, et al. Optimization of ex vivo pressure mediated delivery of antisense oligodeoxynucleotides to ICAM-1 reduces reperfusion injury in rat cardiac allografts. Transplantation 2000;69:1067-74. [90] Katz SM, Bennett F, Stecker K, et al. ICAM-1 antisense oligodeoxynucleotide improves islet allograft survival and function. Cell Transplant 2000;9:817-28.
[91] Vassalli G, Simeoni E, Li JP, Fleury S. Lentiviral gene transfer of the chemokine antagonist RANTES 9-68 prolongs heart graft survival. Transplantation 2006;81:240-6. [92] Fleury S, Li J, Simeoni E, et al. Gene transfer of RANTES and MCP1 chemokine antagonists prolongs cardiac allograft survival. Gene Ther 2006;13:1104-9. [93] 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;7:575-82. [94] 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;116:386-96. [95] Beutelspacher SC, Pillai R, Watson MP, et al. Function of indoleamine 2,3-dioxygenase in corneal allograft rejection and prolongation of allograft survival by over-expression. Eur J Immunol 2006;36:690-700. [96] Liu H, Liu L, Fletcher BS, Visner GA. Novel action of indoleamine 2,3-dioxygenase attenuating acute lung allograft injury. Am J Respir Crit Care Med 2006;173:566-72. [97] Liu H, Liu L, Fletcher BS, Visner GA. Sleeping Beauty–based gene therapy with indoleamine 2,3-dioxygenase inhibits lung allograft fibrosis. Faseb J 2006;20:2384-6. [98] Laurence JM, Wang C, Zheng M, et al. Overexpression of indoleamine dioxygenase in rat liver allografts using a high-efficiency adeno-associated virus vector does not prevent acute rejection. Liver Transpl 2009;15:233-41. [99] Iwata A, Sai S, Moore M, et al. Gene therapy of transplant arteriopathy by liposome-mediated transfection of endothelial nitric oxide synthase. J Heart Lung Transplant 2000;19:1017-28. [100] Lee PC, Wang ZL, Qian S, et al. Endothelial nitric oxide synthase protects aortic allografts from the development of transplant arteriosclerosis. Transplantation 2000;69:1186-92. [101] Shears LL, Kawaharada N, Tzeng E, et al. Inducible nitric oxide synthase suppresses the development of allograft arteriosclerosis. J Clin Invest 1997;100:2035-42. [102] Braudeau C, Bouchet D, Tesson L, et al. Induction of long-term cardiac allograft survival by heme oxygenase-1 gene transfer. Gene Ther 2004;11:701-10. [103] Amersi F, Buelow R, Kato H, et al. Upregulation of heme oxygenase1 protects genetically fat Zucker rat livers from ischemia/reperfusion injury. J Clin Invest 1999;104:1631-9. [104] Coito AJ, Buelow R, Shen XD, et al. Heme oxygenase-1 gene transfer inhibits inducible nitric oxide synthase expression and protects genetically fat Zucker rat livers from ischemia-reperfusion injury. Transplantation 2002;74:96-102. [105] Kato H, Amersi F, Buelow R, et al. Heme oxygenase-1 overexpression protects rat livers from ischemia/reperfusion injury with extended cold preservation. Am J Transplant 2001;1:121-8. [106] Ke B, Buelow R, Shen XD, et al. Heme oxygenase 1 gene transfer prevents CD95/Fas ligand-mediated apoptosis and improves liver allograft survival via carbon monoxide signaling pathway. Hum Gene Ther 2002;13:1189-99. [107] Ke B, Shen XD, Melinek J, et al. Heme oxygenase-1 gene therapy: a novel immunomodulatory approach in liver allograft recipients? Transplant Proc 2001;33:581-2. [108] Li YX, Li G, Dong WP, Lu DR, Tan JM. Protection of human islets from induction of apoptosis and improved islet function with HO-1 gene transduction. Chin Med J (Engl) 2006;119:1639-45. [109] Bouche D, Chauveau C, Roussel JC, et al. Inhibition of graft arteriosclerosis development in rat aortas following heme oxygenase1 gene transfer. Transpl Immunol 2002;9:235-8. [110] Lehmann TG, Wheeler MD, Schoonhoven R, Bunzendahl H, Samulski RJ, Thurman RG. Delivery of Cu/Zn-superoxide dismutase genes with a viral vector minimizes liver injury and improves survival after liver transplantation in the rat. Transplantation 2000;69:1051-7.
J.M. Laurence et al. / Transplantation Reviews 23 (2009) 159–170 [111] Lehmann TG, Wheeler MD, Schwabe RF, et al. Gene delivery of Cu/Zn-superoxide dismutase improves graft function after transplantation of fatty livers in the rat. Hepatology 2000;32: 1255-64. [112] Benhamou PY, Moriscot C, Richard MJ, et al. Adenovirus-mediated catalase gene transfer reduces oxidant stress in human, porcine and rat pancreatic islets. Diabetologia 1998;41:1093-100. [113] He SQ, Zhang YH, Venugopal SK, et al. Delivery of antioxidative enzyme genes protects against ischemia/reperfusion-induced liver injury in mice. Liver Transpl 2006;12:1869-79. [114] Swenson KM, Ke B, Wang T, et al. Fas ligand gene transfer to renal allografts in rats: effects on allograft survival. Transplantation 1998; 65:155-60. [115] Lau HT, Yu M, Fontana A, Stoeckert Jr CJ. Prevention of islet allograft rejection with engineered myoblasts expressing FasL in mice. Science 1996;273:109-12. [116] Allison J, Georgiou HM, Strasser A, Vaux DL. Transgenic expression of CD95 ligand on islet beta cells induces a granulocytic infiltration but does not confer immune privilege upon islet allografts. Proc Natl Acad Sci U S A 1997;94:3943-7. [117] 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;3:738-43. [118] Takeuchi T, Ueki T, Nishimatsu H, et al. Accelerated rejection of Fas ligand–expressing heart grafts. J Immunol 1999;162:518-22. [119] Li XK, Okuyama T, Tamura A, et al. Prolonged survival of rat liver allografts transfected with Fas ligand–expressing plasmid. Transplantation 1998;66:1416-23. [120] Grey ST, Arvelo MB, Hasenkamp W, Bach FH, Ferran C. A20 inhibits cytokine-induced apoptosis and nuclear factor kappaBdependent gene activation in islets. J Exp Med 1999;190:1135-46. [121] 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: 543-53. [122] Kunter U, Daniel S, Arvelo MB, et al. Combined expression of A1 and A20 achieves optimal protection of renal proximal tubular epithelial cells. Kidney Int 2005;68:1520-32. [123] 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;67:775-83. [124] Barcia RN, Dana MR, Kazlauskas A. Corneal graft rejection is accompanied by apoptosis of the endothelium and is prevented by gene therapy with Bcl-xL. Am J Transplant 2007;7:2082-9. [125] Grey ST, Longo C, Shukri T, et al. Genetic engineering of a suboptimal islet graft with A20 preserves beta cell mass and function. J Immunol 2003;170:6250-6. [126] Sykes M. Mixed chimerism and transplant tolerance. Immunity 2001; 14:417-24. [127] 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;55:197-202. [128] 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;154:1587-95. [129] Mayfield RS, Hayashi H, Sawada T, et al. The mechanism of specific prolongation of class I–mismatched skin grafts induced by retroviral gene therapy. Eur J Immunol 1997;27:1177-81. [130] Fry JW, Morris PJ, Wood KJ. Adenoviral transfer of a single donorspecific MHC class I gene to recipient bone marrow cells can induce specific immunological unresponsiveness in vivo. Gene Ther 2002; 9:220-6. [131] 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;159:152-8.
169
[132] 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;57:990-6. [133] Graeb C, Justl M, Scherer MN, et al. Use of an adenoviral vector to express soluble donor-major histocompatibility complex molecules capable of suppressing the immune response in rat transplant recipients. Hum Immunol 2002;63:844-52. [134] 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;64:1414-23. [135] Sonntag KC, Emery DW, Yasumoto A, et al. Tolerance to solid organ transplants through transfer of MHC class II genes. J Clin Invest 2001;107:65-71. [136] Zhang JL, Cai J, Walls S, et al. Tolerance by selective in vivo expansion of foreign major histocompatibility complex–transduced autologous bone marrow. Transplantation 2005;80:362-9. [137] Hawiger D, Inaba K, Dorsett Y, et al. Dendritic cells induce peripheral T cell unresponsiveness under steady state conditions in vivo. J Exp Med 2001;194:769-79. [138] Grohmann U, Bianchi R, Orabona C, et al. Functional plasticity of dendritic cell subsets as mediated by CD40 versus B7 activation. J Immunol 2003;171:2581-7. [139] Takayama T, Morelli AE, Robbins PD, Tahara H, Thomson AW. Feasibility of CTLA4Ig gene delivery and expression in vivo using retrovirally transduced myeloid dendritic cells that induce alloantigen-specific T cell anergy in vitro. Gene Ther 2000;7: 1265-73. [140] 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; 66:1567-74. [141] Funeshima N, Fujino M, Kitazawa Y, et al. Inhibition of allogeneic Tcell responses by dendritic cells expressing transduced indoleamine 2,3-dioxygenase. J Gene Med 2005;7:565-75. [142] Zhang HG, Su X, Liu D, et al. Induction of specific T cell tolerance by Fas ligand–expressing antigen-presenting cells. J Immunol 1999;162: 1423-30. [143] 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; 66:1810-7. [144] Zhu M, Wei MF, Liu F, Shi HF, Wang G. Interleukin-10 modified dendritic cells induce allo-hyporesponsiveness and prolong small intestine allograft survival. World J Gastroenterol 2003;9:2509-12. [145] Min WP, Gorczynski R, Huang XY, et al. Dendritic cells genetically engineered to express Fas ligand induce donor-specific hyporesponsiveness and prolong allograft survival. J Immunol 2000;164:161-7. [146] O'Rourke RW, Kang SM, Lower JA, et al. A dendritic cell line genetically modified to express CTLA4-IG as a means to prolong islet allograft survival. Transplantation 2000;69:1440-6. [147] Kaneko K, Wang Z, Kim SH, Morelli AE, Robbins PD, Thomson AW. Dendritic cells genetically engineered to express IL-4 exhibit enhanced IL-12p70 production in response to CD40 ligation and accelerate organ allograft rejection. Gene Ther 2003;10:143-52. [148] Wood KJ, Sakaguchi S. Regulatory T cells in transplantation tolerance. Nat Rev Immunol 2003;3:199-210. [149] Chai JG, Xue SA, Coe D, et al. Regulatory T cells, derived from naïve CD4+CD25− T cells by in vitro FoxP3 gene transfer, can induce transplantation tolerance. Transplantation 2005;79:1310-6. [150] Tsang JYS, Tanriver Y, Jiang S, et al. Conferring indirect allospecificity on CD4+CD25+T regs by TCR gene transfer favors transplantation tolerance in mice. J Clin Invest 2008;118: 3619-28.
170
J.M. Laurence et al. / Transplantation Reviews 23 (2009) 159–170
[151] Relph K, Harrington K, Pandha H. Recent developments and current status of gene therapy using viral vectors in the United Kingdom. BMJ 2004;329:839-42. [152] Raper SE, Chirmule N, Lee FS, et al. Fatal systemic inflammatory response syndrome in a ornithine transcarbamylase deficient patient following adenoviral gene transfer. Mol Genet Metab 2003; 80:148-58. [153] Hacein-Bey-Abina S, von Kalle C, Schmidt M, et al. A serious adverse event after successful gene therapy for X-linked severe combined immunodeficiency. N Engl J Med 2003;348:255-6. [154] Hacein-Bey-Abina S, Garrigue A, Wang GP, et al. Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J Clin Invest 2008;118:3132-42. [155] Howe SJ, Mansour MR, Schwartzwaelder K, et al. Insertional mutagenesis combined with acquired somatic mutations causes leukemogenesis following gene therapy of SCID-X1 patients. J Clin Invest 2008;118:3143-50.
[156] Paredes D, Gambra MP, Cervera C, et al. Characterization of the organ donor with bacteremia. Transplant Proc 2007;39:2083-5. [157] Wang Z, Ma HI, Li J, Sun L, Zhang J, Xiao X. Rapid and highly efficient transduction by double-stranded adeno-associated virus vectors in vitro and in vivo. Gene Ther 2003;10:2105-11. [158] Nathwani AC, Gray JT, Ng CY, et al. Self-complementary adenoassociated virus vectors containing a novel liver-specific human factor IX expression cassette enable highly efficient transduction of murine and nonhuman primate liver. Blood 2006;107: 2653-61. [159] Muruve DA, Barnes MJ, Stillman IE, Libermann TA. Adenoviral gene therapy leads to rapid induction of multiple chemokines and acute neutrophil-dependent hepatic injury in vivo. Hum Gene Ther 1999;10:965-76. [160] 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 Pt 1):708-22.